EP3319985B1 - Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers - Google Patents

Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers Download PDF

Info

Publication number
EP3319985B1
EP3319985B1 EP16736073.4A EP16736073A EP3319985B1 EP 3319985 B1 EP3319985 B1 EP 3319985B1 EP 16736073 A EP16736073 A EP 16736073A EP 3319985 B1 EP3319985 B1 EP 3319985B1
Authority
EP
European Patent Office
Prior art keywords
cancer
peptide
cell
cells
peptides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP16736073.4A
Other languages
German (de)
French (fr)
Other versions
EP3319985A2 (en
Inventor
Andrea Mahr
Toni Weinschenk
Colette SONG
Oliver Schoor
Jens Fritsche
Harpreet Singh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immatics Biotechnologies GmbH
Original Assignee
Immatics Biotechnologies GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immatics Biotechnologies GmbH filed Critical Immatics Biotechnologies GmbH
Priority to EP20175260.7A priority Critical patent/EP3736283A1/en
Priority to SI201631018T priority patent/SI3319985T1/en
Priority to EP23216397.2A priority patent/EP4321172A2/en
Priority to RS20201531A priority patent/RS61203B1/en
Priority to PL16736073T priority patent/PL3319985T3/en
Publication of EP3319985A2 publication Critical patent/EP3319985A2/en
Application granted granted Critical
Publication of EP3319985B1 publication Critical patent/EP3319985B1/en
Priority to HRP20210076TT priority patent/HRP20210076T1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001152Transcription factors, e.g. SOX or c-MYC
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/635Parathyroid hormone, i.e. parathormone; Parathyroid hormone-related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/16Aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • Squamous cell carcinoma and adenocarcinoma represent the two most common subtypes of esophageal cancer. Both subtypes are more common in men than in women, but they display distinct geographical distributions. Squamous cell carcinoma is more prevalent in low-resource regions with particularly high incidence rates in the Islamic Republic of Iran, parts of China and clouds. Adenocarcinoma is the most common type of esophageal cancer among Caucasians and populations with a high socioeconomic status, with the United Kingdom, Australia, the Netherlands and the USA leading the way.
  • Patients with HER2-positive tumors should be treated according to the guidelines for gastric cancer using a combination of cisplatin, fluorouracil and trastuzumab, as randomized data for targeted therapies in esophageal cancer are very limited (Stahl et al., 2013; Leitline Magenkarzinom, 2012).
  • HLA class II molecules Since the constitutive expression of HLA class II molecules is usually limited to immune cells, the possibility of isolating class II peptides directly from primary tumors was previously not considered possible. However, Dengjel et al. were successful in identifying a number of MHC Class II epitopes directly from tumors ( WO 2007/028574 , EP 1 760 088 B1 ).
  • CD8 and CD4 dependent Since both types of response, CD8 and CD4 dependent, contribute jointly and synergistically to the anti-tumor effect, the identification and characterization of tumor-associated antigens recognized by either CD8+ T cells (ligand: MHC class I molecule + peptide epitope) or by CD4-positive T-helper cells (ligand: MHC class II molecule + peptide epitope) is important in the development of tumor vaccines.
  • MHC-class-I-binding peptides are usually 8-12 amino acid residues in length and usually contain two conserved residues ("anchors") in their sequence that interact with the corresponding binding groove of the MHC-molecule. In this way each MHC allele has a "binding motif" determining which peptides can bind specifically to the binding groove.
  • peptides In the MHC class I dependent immune reaction, peptides not only have to be able to bind to certain MHC class I molecules expressed by tumor cells, they subsequently also have to be recognized by T cells bearing specific T cell receptors (TCR).
  • TCR T cell receptors
  • the immunogenicity of the underlying peptides is secondary. In these cases, the presentation is the determining factor.
  • Rizzolo et al. (in: Conventional and microwave-assisted SPPS approach: a comparative synthesis of PTHrP(1-34)NH2D3 J Pept Sci. 2011 Oct;17(10):708-14 ) relates to a synthesis method for polypeptides, and compares Fmoc/tBu MW-assisted SPPS of 1-34 N-terminal fragment of parathyroid hormone-related peptide (PTHrP) with its conventional SPPS carried out at RT. During the stepwise elongation of the resin-bound peptide, monitoring was conducted by performing MW-assisted mini-cleavages and analyzing them by UPLC-ESI-MS.
  • PTHrP parathyroid hormone-related peptide
  • the present invention relates to a peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  • SEQ ID NO: 9 is for use according to the invention SEQ ID No Sequence Gene ID(s) Official Gene Symbol(s) 1 STYGGGLSV 3861, 3868 KRT14, KRT16 2 SLYNLGGSKRISI 3852 KRT5 3 TASAITPSV 3852 KRT5 4 ALFGTILEL 2769 GNA15 5 NLMASQPQL 5317 PKP1 6 LLSGDLIFL 2709 GJB5 7 SIFEGLLSGV 2709 GJB5 8 ALLDGGSEAYWRV 84985 FAM83A 9 HLIAEIHTA 5744 PTHLH 10 SLDENSDQQV 6273 S100A2 11 ALWLPTDSATV 3914 LAMB3 12 GLASRILDA 3914 LAMB3 13 SLSPVILGV 26525 IL36RN 14 RLPNAGTQV 3655 ITGA6 15 LLANGVYAA 55107 ANO1 16 VLAEGGEGV 10630 PDPN 17 MISRTPEV 2155, 28
  • peptides - alone or in combination - from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 93. More preferred are the peptides - alone or in combination - selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 76 (see Table 1), and their uses in the immunotherapy of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia, and preferably esophageal cancer.
  • peptides - alone or in combination - selected from the group consisting of SEQ ID No. 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, 19, 25, 26, 30, 32, 34, 37, 40, 51, 55, 57, 58, 59, 62, 81, and 82, and their uses in the immunotherapy of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia, and preferably esophageal cancer. Further particularly preferred is the peptide for use according to SEQ ID NO: 9.
  • Table 4A Peptides as disclosed and their specific uses in other proliferative diseases, especially in other cancerous diseases.
  • SEQ ID NO: 9 is for use according to the invention. The table shows for selected peptides on which additional tumor types they were found and either over-presented on more than 5% of the measured tumor samples, or presented on more than 5% of the measured tumor samples with a ratio of geometric means tumor vs normal tissues being larger than 3. Over-presentation is defined as higher presentation on the tumor sample as compared to the normal sample with highest presentation.
  • adipose tissue adrenal gland, artery, bone marrow, brain, central nerve, colon, duodenum, esophagus, gallbladder, heart, kidney, liver, lung, lymph node, mononuclear white blood cells, pancreas, peripheral nerve, peritoneum, pituitary, pleura, rectum, salivary gland, skeletal muscle, skin, small intestine, spleen, stomach, thymus, thyroid gland, trachea, ureter, urinary bladder, vein.
  • the table shows, like Table 4A, for selected peptides on which additional tumor types they were found showing over-presentation (including specific presentation) on more than 5% of the measured tumor samples, or presentation on more than 5% of the measured tumor samples with a ratio of geometric means tumor vs normal tissues being larger than 3.
  • Over-presentation is defined as higher presentation on the tumor sample as compared to the normal sample with highest presentation.
  • another aspect of the present invention relates to the use of the peptides according to the present invention for the - preferably combined - treatment of a proliferative disease selected from the group of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • a proliferative disease selected from the group of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • the present invention further relates to an expression vector expressing a nucleic acid according to the present invention.
  • the present invention further relates to T-cell receptors (TCRs), in particular soluble TCR (sTCRs) and cloned TCRs engineered into autologous or allogeneic T cells, and methods of making these, as well as NK cells or other cells bearing said TCR or cross-reacting with said TCRs.
  • TCRs T-cell receptors
  • sTCRs soluble TCR
  • cloned TCRs engineered into autologous or allogeneic T cells
  • the antibodies and TCRs are additional embodiments of the immunotherapeutic use of the peptides according to the invention at hand.
  • the present invention further relates to a host cell comprising a nucleic acid according to the present invention or an expression vector as described before.
  • the present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably is a dendritic cell.
  • PTHLH up-regulation was shown to be associated with poor pathologic differentiation and poor prognosis in patients with head and neck squamous cell carcinoma (Lv et al., 2014).
  • PTHLH was shown to be up-regulated through p38 MAPK signaling, which contributes to colon cancer cell extravasation of the lung by caspase-independent death in endothelial cells of the lung microsvasculature (Urosevic et al., 2014).
  • PTHLH was shown to be significantly differentially expressed in squamous cell carcinoma compared with normal skin (Prasad et al., 2014).
  • PTHLH was shown to positively modulate cell cycle progression and to change the expression of proteins involved in cell cycle regulation via ERK1/2, p38, MAPK, and PI3K signaling pathways in the colorectal adenocarcinoma cell line Caco-2 (Calvo et al., 2014).
  • T-cell response means the specific proliferation and activation of effector functions induced by a peptide in vitro or in vivo.
  • effector functions may be lysis of peptide-pulsed, peptide-precursor pulsed or naturally peptide-presenting target cells, secretion of cytokines, preferably Interferon-gamma, TNF-alpha, or IL-2 induced by peptide, secretion of effector molecules, preferably granzymes or perforins induced by peptide, or degranulation.
  • polypeptide designates a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids.
  • the length of the polypeptide is not critical to the invention as long as the correct epitopes are maintained.
  • polypeptide is meant to refer to molecules containing more than about 30 amino acid residues.
  • expression product means the polypeptide or protein that is the natural translation product of the gene and any nucleic acid sequence coding equivalents resulting from genetic code degeneracy and thus coding for the same amino acid(s).
  • portion when used in relation to polypeptides, refer to a continuous sequence of residues, such as amino acid residues, which sequence forms a subset of a larger sequence.
  • residues such as amino acid residues
  • oligopeptides resulting from such treatment would represent portions, segments or fragments of the starting polypeptide.
  • these terms refer to the products produced by treatment of said polynucleotides with any of the endonucleases.
  • the peptide for use is part of a fusion protein which comprises the 80 N-terminal amino acids of the HLA-DR antigen-associated invariant chain (p33, in the following "li") as derived from the NCBI, GenBank Accession number X00497.
  • the peptides for use of the present invention can be fused to an antibody as described herein, or a functional part thereof, in particular into a sequence of an antibody, so as to be specifically targeted by said antibody, or, for example, to or into an antibody that is specific for dendritic cells as described herein.
  • Peptides comprising the sequences described above may be synthesized with additional chemical groups present at their amino and/or carboxy termini, to enhance the stability, bioavailability, and/or affinity of the peptides.
  • additional chemical groups such as carbobenzoxyl, dansyl, or t-butyloxycarbonyl groups may be added to the peptides' amino termini.
  • an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the peptides' amino termini.
  • the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the peptides' carboxy termini.
  • peptides (at least those containing peptide linkages between amino acid residues) may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lukas et al. (Lukas et al., 1981) and by references as cited therein.
  • Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide.
  • peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50 % scavenger mix.
  • Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible (see, for example, (Bruckdorfer et al., 2004), and the references as cited therein).
  • the discovery pipeline XPRESIDENT® v2. allows for a direct absolute quantitation of MHC-, preferably HLA-restricted, peptide levels on cancer or other infected tissues. Briefly, the total cell count was calculated from the total DNA content of the analyzed tissue sample. The total peptide amount for a TUMAP in a tissue sample was measured by nanoLC-MS/MS as the ratio of the natural TUMAP and a known amount of an isotope-labelled version of the TUMAP, the so-called internal standard.
  • the efficiency of TUMAP isolation was determined by spiking peptide:MHC complexes of all selected TUMAPs into the tissue lysate at the earliest possible point of the TUMAP isolation procedure and their detection by nanoLC-MS/MS following completion of the peptide isolation procedure.
  • the total cell count and the amount of total peptide were calculated from triplicate measurements per tissue sample.
  • the peptide-specific isolation efficiencies were calculated as an average from 10 spike experiments each measured as a triplicate (see Example 6 and Table 12).
  • T-cell receptor refers to a heterodimeric molecule comprising an alpha polypeptide chain (alpha chain) and a beta polypeptide chain (beta chain), wherein the heterodimeric receptor is capable of binding to a peptide antigen presented by an HLA molecule.
  • the term also includes so-called gamma/delta TCRs.
  • the description in another aspect relates to methods according to the description, wherein the antigen is loaded onto class I MHC molecules expressed on the surface of a suitable antigen-presenting cell or artificial antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell or the antigen is loaded onto class I MHC tetramers by tetramerizing the antigen/class I MHC complex monomers.
  • TCR gamma variable domain refers to the concatenation of the TCR gamma V (TRGV) region without leader region (L), and the TCR gamma J (TRGJ) region
  • TCR gamma constant domain refers to the extracellular TRGC region, or to a C-terminal truncated TRGC sequence.
  • TCRs of the present description preferably bind to a peptide-HLA molecule complex with a binding affinity (KD) of about 100 ⁇ M or less, about 50 ⁇ M or less, about 25 ⁇ M or less, or about 10 ⁇ M or less. More preferred are high affinity TCRs having binding affinities of about 1 ⁇ M or less, about 100 nM or less, about 50 nM or less, about 25 nM or less.
  • KD binding affinity
  • Non-limiting examples of preferred binding affinity ranges for TCRs of the present invention include about 1 nM to about 10 nM; about 10 nM to about 20 nM; about 20 nM to about 30 nM; about 30 nM to about 40 nM; about 40 nM to about 50 nM; about 50 nM to about 60 nM; about 60 nM to about 70 nM; about 70 nM to about 80 nM; about 80 nM to about 90 nM; and about 90 nM to about 100 nM.
  • alpha/beta heterodimeric TCRs of the present description may have a TRAC constant domain sequence and a TRBC1 or TRBC2 constant domain sequence, and the TRAC constant domain sequence and the TRBC1 or TRBC2 constant domain sequence of the TCR may be linked by the native disulfide bond between Cys4 of exon 2 of TRAC and Cys2 of exon 2 of TRBC1 or TRBC2.
  • TCRs of the present description may comprise a detectable label selected from the group consisting of a radionuclide, a fluorophore and biotin. TCRs of the present description may be conjugated to a therapeutically active agent, such as a radionuclide, a chemotherapeutic agent, or a toxin.
  • a therapeutically active agent such as a radionuclide, a chemotherapeutic agent, or a toxin.
  • a TCR comprising at least one mutation in the TCR alpha chain and/or TCR beta chain has a binding affinity for, and/or a binding half-life for, a peptide-HLA molecule complex, which is at least double that of a TCR comprising the non-mutated TCR alpha chain and/or non-mutated TCR beta chain.
  • Affinity-enhancement of tumor-specific TCRs, and its exploitation, relies on the existence of a window for optimal TCR affinities.
  • nucleic acids encoding TCRs of the present description may be operably linked to strong promoters, such as retroviral long terminal repeats (LTRs), cytomegalovirus (CMV), murine stem cell virus (MSCV) U3, phosphoglycerate kinase (PGK), ⁇ -actin, ubiquitin, and a simian virus 40 (SV40)/CD43 composite promoter, elongation factor (EF)-1a and the spleen focus-forming virus (SFFV) promoter.
  • promoter is heterologous to the nucleic acid being expressed.
  • the alpha and beta chains of a TCR of the present invention may be encoded by nucleic acids located in separate vectors, or may be encoded by polynucleotides located in the same vector.
  • TCR-alpha and TCR-beta chains of the introduced TCR be transcribed at high levels.
  • the TCR-alpha and TCR-beta chains of the present description may be cloned into bi-cistronic constructs in a single vector, which has been shown to be capable of overcoming this obstacle.
  • TCR-alpha and TCR-beta chains are used to coordinate expression of both chains, because the TCR-alpha and TCR-beta chains are generated from a single transcript that is broken into two proteins during translation, ensuring that an equal molar ratio of TCR-alpha and TCR-beta chains are produced.
  • IRS intra-ribosomal entry site
  • Nucleic acids encoding TCRs of the present description may be codon optimized to increase expression from a host cell. Redundancy in the genetic code allows some amino acids to be encoded by more than one codon, but certain codons are less "optimal” than others because of the relative availability of matching tRNAs as well as other factors (Gustafsson et al., 2004). Modifying the TCR-alpha and TCR-beta gene sequences such that each amino acid is encoded by the optimal codon for mammalian gene expression, as well as eliminating mRNA instability motifs or cryptic splice sites, has been shown to significantly enhance TCR-alpha and TCR-beta gene expression (Scholten et al., 2006).
  • compositions comprise the peptides either in the free form or in the form of a pharmaceutically acceptable salt (see also above).
  • a pharmaceutically acceptable salt refers to a derivative of the disclosed peptides wherein the peptide is modified by making acid or base salts of the agent.
  • acid salts are prepared from the free base (typically wherein the neutral form of the drug has a neutral -NH2 group) involving reaction with a suitable acid.
  • Suitable acids for preparing acid salts include both organic acids, e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methane sulfonic acid, ethane sulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid phosphoric acid and the like.
  • preparation of basic salts of acid moieties which may be present on a peptide are prepared using a pharmaceutically acceptable base such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine or the like.
  • the medicament for use of the present invention is an immunotherapeutics such as a vaccine. It may be administered directly into the patient, into the affected organ or systemically i.d., i.m., s.c., i.p. and i.v., or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation of immune cells derived from the patient, which are then re-administered to the patient. If the nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2.
  • cytokines such as interleukin-2.
  • the peptide may be substantially pure, or combined with an immune-stimulating adjuvant (see below) or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, for example liposomes.
  • the peptide may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see WO 95/18145 and (Longenecker et al., 1993)).
  • KLH keyhole limpet haemocyanin
  • mannan see WO 95/18145 and (Longenecker et al., 1993)
  • the peptide may also be tagged, may be a fusion protein, or may be a hybrid molecule.
  • the peptides whose sequence is given in the present invention are expected to stimulate CD4 or CD8 T cells.
  • a further aspect of the invention provides a nucleic acid (for example a polynucleotide) encoding a peptide for use of the invention.
  • the polynucleotide may be, for example, DNA, cDNA, PNA, RNA or combinations thereof, either single- and/or double-stranded, or native or stabilized forms of polynucleotides, such as, for example, polynucleotides with a phosphorothioate backbone and it may or may not contain introns so long as it codes for the peptide.
  • a still further aspect of the invention provides an expression vector expressing a peptide for use according to the invention.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc. New Haven, CN, USA.
  • a desirable method of modifying the DNA encoding the polypeptide of the invention employs the polymerase chain reaction as disclosed by Saiki RK, et al. (Saiki et al., 1988).This method may be used for introducing the DNA into a suitable vector, for example by engineering in suitable restriction sites, or it may be used to modify the DNA in other useful ways as is known in the art. If viral vectors are used, pox- or adenovirus vectors are preferred.
  • the DNA (or in the case of retroviral vectors, RNA) may then be expressed in a suitable host to produce a polypeptide comprising the peptide or variant of the invention.
  • RNA Ribonucleic acid
  • the DNA encoding the peptide for use of the invention may be used in accordance with known techniques, appropriately modified in view of the teachings contained herein, to construct an expression vector, which is then used to transform an appropriate host cell for the expression and production of the polypeptide of the invention.
  • Such techniques include those disclosed, for example, in US 4,440,859 , 4,530,901 , 4,582,800 , 4,677,063 , 4,678,751 , 4,704,362 , 4,710,463 , 4,757,006 , 4,766,075 , and 4,810,648 .
  • a typical mammalian cell vector plasmid for constitutive expression comprises the CMV or SV40 promoter with a suitable poly A tail and a resistance marker, such as neomycin.
  • a suitable poly A tail and a resistance marker, such as neomycin.
  • pSVL available from Pharmacia, Piscataway, NJ, USA.
  • An example of an inducible mammalian expression vector is pMSG, also available from Pharmacia.
  • Useful yeast plasmid vectors are pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
  • CMV human cytomegalovirus
  • the strong human cytomegalovirus (CMV) promoter regulatory region drives constitutive protein expression levels as high as 1 mg/L in COS cells. For less potent cell lines, protein levels are typically ⁇ 0.1 mg/L.
  • the presence of the SV40 replication origin will result in high levels of DNA replication in SV40 replication permissive COS cells.
  • CMV vectors for example, can contain the pMB1 (derivative of pBR322) origin for replication in bacterial cells, the b-lactamase gene for ampicillin resistance selection in bacteria, hGH polyA, and the f1 origin.
  • the present invention also relates to a host cell transformed with a polynucleotide vector construct of the present invention.
  • the host cell can be either prokaryotic or eukaryotic.
  • Bacterial cells may be preferred prokaryotic host cells in some circumstances and typically are a strain of E. coli such as, for example, the E. coli strains DH5 available from Bethesda Research Laboratories Inc., Bethesda, MD, USA, and RR1 available from the American Type Culture Collection (ATCC) of Rockville, MD, USA (No ATCC 31343).
  • ATCC American Type Culture Collection
  • Successfully transformed cells i.e. cells that contain a DNA construct of the present invention, can be identified by well-known techniques such as PCR. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
  • host cells of the invention are useful in the preparation of the peptide for use of the invention, for example bacterial, yeast and insect cells.
  • other host cells may be useful in certain therapeutic methods.
  • antigen-presenting cells such as dendritic cells, may usefully be used to express the peptide for use of the invention such that they may be loaded into appropriate MHC molecules.
  • the current invention provides a host cell comprising a nucleic acid or an expression vector according to the invention.
  • a further aspect of the invention provides a method of producing a peptide for use, the method comprising culturing a host cell and isolating the peptide from the host cell or its culture medium.
  • the medicament for use of the invention may also include one or more adjuvants.
  • adjuvants are substances that non-specifically enhance or potentiate the immune response (e.g., immune responses mediated by CD8-positive T cells and helper-T (TH) cells to an antigen, and would thus be considered useful in the medicament of the present invention.
  • Suitable adjuvants include, but are not limited to, 1018 ISS, aluminum salts, AMPLIVAX®, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, flagellin or TLR5 ligands derived from flagellin, FLT3 ligand, GM-CSF, IC30, IC31, Imiquimod (ALDARA®), resiquimod, ImuFact IMP321, Interleukins as IL-2, IL-13, IL-21, Interferon-alpha or -beta, or pegylated derivatives thereof, IS Patch, ISS, ISCOMATRIX, ISCOMs, JuvImmune®, LipoVac, MALP2, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, water-in-oil and oil-in-water emulsions, OK
  • Adjuvants such as Freund's or GM-CSF are preferred.
  • Several immunological adjuvants e.g., MF59
  • cytokines may be used.
  • cytokines have been directly linked to influencing dendritic cell migration to lymphoid tissues (e.g., TNF-), accelerating the maturation of dendritic cells into efficient antigen-presenting cells for T-lymphocytes (e.g., GM-CSF, IL-1 and IL-4) ( U.S. Pat. No.
  • immunoadjuvants e.g., IL-12, IL-15, IL-23, IL-7, IFN-alpha. IFN-beta
  • IL-12, IL-15, IL-23, IL-7, IFN-alpha. IFN-beta e.g., IL-12, IL-15, IL-23, IL-7, IFN-alpha. IFN-beta
  • a CpG TLR9 antagonist is dSLIM (double Stem Loop Immunomodulator) by Mologen (Berlin, Germany) which is a preferred component of the pharmaceutical composition of the present invention.
  • TLR binding molecules such as RNA binding TLR 7, TLR 8 and/or TLR 9 may also be used.
  • CpGs e.g. CpR, Idera
  • dsRNA analogues such as Poly(I:C) and derivates thereof (e.g. AmpliGen®, Hiltonol®, poly-(ICLC), poly(IC-R), poly(I:C12U), non-CpG bacterial DNA or RNA as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitinib, Bevacizumab®, celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafenib, temozolomide, temsirolimus, XL-999, CP-547632, pazopanib, VEGF Trap, ZD2171, AZD2171, anti-CTLA4, other antibodies targeting key structures of the immune system (e.g.
  • anti-CD40, anti-TGFbeta, anti-TNFalpha receptor) and SC58175, which may act therapeutically and/or as an adjuvant may act therapeutically and/or as an adjuvant.
  • concentrations of adjuvants and additives useful in the context of the present invention can readily be determined by the skilled artisan without undue experimentation.
  • Preferred adjuvants are anti-CD40, imiquimod, resiquimod, GM-CSF, cyclophosphamide, sunitinib, bevacizumab, interferon-alpha, CpG oligonucleotides and derivates, poly-(I:C) and derivates, RNA, sildenafil, and particulate formulations with PLG or virosomes.
  • the pharmaceutical composition for use according to the invention is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod, resiquimod, and interferon-alpha.
  • colony-stimulating factors such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod, resiquimod, and interferon-alpha.
  • the pharmaceutical composition for use according to the invention is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod and resiquimod.
  • the adjuvant is cyclophosphamide, imiquimod or resiquimod.
  • Even more preferred adjuvants are Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, poly-ICLC (Hiltonol®) and anti-CD40 mAB, or combinations thereof.
  • composition is used for parenteral administration, such as subcutaneous, intradermal, intramuscular or oral administration.
  • parenteral administration such as subcutaneous, intradermal, intramuscular or oral administration.
  • the peptides and optionally other molecules are dissolved or suspended in a pharmaceutically acceptable, preferably aqueous carrier.
  • the composition can contain excipients, such as buffers, binding agents, blasting agents, diluents, flavors, lubricants, etc.
  • the peptides can also be administered together with immune stimulating substances, such as cytokines.
  • An extensive listing of excipients that can be used in such a composition can be, for example, taken from A. Kibbe, Handbook of Pharmaceutical Excipients (Kibbe, 2000 ).
  • the composition can be used for a prevention, prophylaxis and/or therapy of adenomatous or cancerous diseases. Exemplary formulations can be found in, for example, EP2112253 .
  • the immune response triggered by the vaccine for use according to the invention attacks the cancer in different cell-stages and different stages of development. Furthermore, different cancer associated signaling pathways are attacked. This is an advantage over vaccines that address only one or few targets, which may cause the tumor to easily adapt to the attack (tumor escape). Furthermore, not all individual tumors express the same pattern of antigens. Therefore, a combination of several tumor-associated peptides ensures that every single tumor bears at least some of the targets.
  • the composition is designed in such a way that each tumor is expected to express several of the antigens and cover several independent pathways necessary for tumor growth and maintenance. Thus, the vaccine can easily be used "off-the-shelf" for a larger patient population.
  • the peptide for use of the present invention can be used to generate and develop specific antibodies against MHC/peptide complexes. These can be used for therapy, targeting toxins or radioactive substances to the diseased tissue. Another use of these antibodies can be targeting radionuclides to the diseased tissue for imaging purposes such as PET. This use can help to detect small metastases or to determine the size and precise localization of diseased tissues.
  • MHC human major histocompatibility complex
  • the antibody is binding with a binding affinity of below 20 nanomolar, preferably of below 10 nanomolar, to the complex, which is also regarded as "specific" in the context of the present invention.
  • the present invention relates to peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  • the present invention further relates to the peptide for use according to the invention that has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I.
  • MHC human major histocompatibility complex
  • the present invention further relates to the peptides according to the invention, wherein the peptide is part of a fusion protein, comprising the N-terminal 80 amino acids of the HLA-DR antigen-associated invariant chain (li).
  • Another embodiment of the present invention relates to a non-naturally occurring peptide for use wherein said peptide consists of the amino acid sequence according to SEQ ID No: 9 and has been synthetically produced (e.g. synthesized) as a pharmaceutically acceptable salt.
  • Methods to synthetically produce peptides are well known in the art.
  • the salts of the peptide for use according to the present invention differ substantially from the peptide in its state in vivo, as the peptide as generated in vivo is no salt.
  • the non-natural salt form of the peptide mediates the solubility of the peptide, in particular in the context of pharmaceutical compositions comprising the peptides for use, e.g. the peptide vaccines for use as disclosed herein.
  • the salts are pharmaceutically acceptable salts of the peptide for use.
  • These salts according to the invention include alkaline and earth alkaline salts such as salts of the Hofmeister series comprising as anions PO 4 3- , SO 4 2- , CH 3 COO - , Cl - , Br - , NO 3 - , ClO 4 - , I - , SCN - and as cations NH 4 + , Rb + , K + , Na + , Cs + , Li + , Zn 2+ , Mg 2+ , Ca 2+ , Mn 2+ , Cu 2+ and Ba 2+ .
  • Particularly salts are selected from (NH 4 ) 3 PO 4 , (NH 4 ) 2 HPO 4 , (NH 4 )H 2 PO 4 , (NH 4 ) 2 SO 4 , NH 4 CH 3 COO, NH 4 Cl, NH 4 Br, NH 4 NO 3 , NH 4 ClO 4 , NH 4 I, NH 4 SCN, Rb 3 PO 4 , Rb 2 HPO 4 , RbH 2 PO 4 , Rb 2 SO 4 , Rb 4 CH 3 COO, Rb 4 Cl, Rb 4 Br, Rb 4 NO 3 , Rb 4 ClO 4 , Rb 4 I, Rb 4 SCN, K 3 PO 4 , K 2 HPO 4 , KH 2 PO 4 , K 2 SO 4 , KCH 3 COO, KCl, KBr, KNO 3 , KClO 4 , KI, KSCN, Na 3 PO 4 , Na 2 HPO 4 , NaH 2 PO 4 , Na 2 SO 4 , NaCH
  • peptides (at least those containing peptide linkages between amino acid residues) may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lukas et al. (Lukas et al., 1981) and by references as cited therein.
  • Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide.
  • Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine).
  • glutamine or asparagine are C-terminal residues, use is made of the 4,4'-dimethoxybenzhydryl group for protection of the side chain amido functionalities.
  • the solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalizing agent).
  • the peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N, N-dicyclohexylcarbodiimide/1hydroxybenzotriazole mediated coupling procedure.
  • peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50 % scavenger mix.
  • Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible (see, for example, (Bruckdorfer et al., 2004), and the references as cited therein).
  • Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide.
  • Any scavengers present are removed by a simple extraction procedure which on lyophilization of the aqueous phase affords the crude peptide free of scavengers.
  • Reagents for peptide synthesis are generally available from e.g. Calbiochem-Novabiochem (Nottingham, UK).
  • the present invention further relates to an expression vector expressing a nucleic acid according to the present invention.
  • the present invention further relates to a peptide according to the present invention, a nucleic acid according to the present invention or an expression vector according to the present invention for use in medicine, in particular in the treatment of esophageal cancer.
  • the present invention further relates to a host cell comprising a nucleic acid according to the invention or an expression vector according to the invention.
  • the present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably a dendritic cell.
  • the present invention further relates to a method of producing a peptide for use according to the present invention, said method comprising culturing the host cell according to the present invention, and isolating the peptide from said host cell or its culture medium.
  • the present invention further relates to activated T cells, produced by the method according to the present invention, wherein said T cells selectively recognizes a cell which aberrantly expresses a polypeptide comprising an amino acid sequence for use according to the present invention.
  • the present invention further relates to a use according to the present invention, wherein the medicament is active against cancer.
  • the present invention further relates to a use according to the invention, wherein the medicament is a vaccine.
  • the present invention further relates to a use according to the invention, wherein the medicament is active against cancer.
  • the present invention further relates to a use according to the invention, wherein said cancer cells are esophageal cancer cells or other solid or hematological tumor cells such as lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • said cancer cells are esophageal cancer cells or other solid or hematological tumor cells such as lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • antibody or “antibodies” is used herein in a broad sense and includes both polyclonal and monoclonal antibodies. In addition to intact or “full” immunoglobulin molecules, also included in the term “antibodies” are fragments (e.g.
  • CDRs, Fv, Fab and Fc fragments or polymers of those immunoglobulin molecules and humanized versions of immunoglobulin molecules, as long as they exhibit any of the desired properties (e.g., specific binding of a esophageal cancer marker (poly)peptide, delivery of a toxin to a esophageal cancer cell expressing a cancer marker gene at an increased level, and/or inhibiting the activity of a esophageal cancer marker polypeptide) according to the invention.
  • desired properties e.g., specific binding of a esophageal cancer marker (poly)peptide, delivery of a toxin to a esophageal cancer cell expressing a cancer marker gene at an increased level, and/or inhibiting the activity of a esophageal cancer marker polypeptide
  • the antibodies of the invention may be purchased from commercial sources.
  • the antibodies of the invention may also be generated using well-known methods. The skilled artisan will understand that either full length esophageal cancer marker polypeptides or fragments thereof may be used to generate the antibodies of the invention.
  • a polypeptide to be used for generating an antibody of the invention may be partially or fully purified from a natural source, or may be produced using recombinant DNA techniques.
  • a cDNA encoding a peptide for use according to the present invention can be expressed in prokaryotic cells (e.g., bacteria) or eukaryotic cells (e.g., yeast, insect, or mammalian cells), after which the recombinant protein can be purified and used to generate a monoclonal or polyclonal antibody preparation that specifically bind the esophageal cancer marker polypeptide used to generate the antibody according to the invention.
  • prokaryotic cells e.g., bacteria
  • eukaryotic cells e.g., yeast, insect, or mammalian cells
  • the antibodies may be tested in ELISA assays or, Western blots, immunohistochemical staining of formalin-fixed cancers or frozen tissue sections. After their initial in vitro characterization, antibodies intended for therapeutic or in vivo diagnostic use are tested according to known clinical testing methods.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a substantially homogeneous population of antibodies, i.e.; the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired antagonistic activity ( US 4,816,567 ).
  • Monoclonal antibodies of the invention may be prepared using hybridoma methods.
  • a hybridoma method a mouse or other appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent.
  • the lymphocytes may be immunized in vitro.
  • In vitro methods are also suitable for preparing monovalent antibodies.
  • Digestion of antibodies to produce fragments thereof, particularly Fab fragments can be accomplished using routine techniques known in the art. For instance, digestion can be performed using papain. Examples of papain digestion are described in WO 94/29348 and US 4,342,566 .
  • Papain digestion of antibodies typically produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual Fc fragment. Pepsin treatment yields a F(ab')2 fragment and a pFc' fragment.
  • the antibody fragments can also include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the non-modified antibody or antibody fragment. These modifications can provide for some additional property, such as to remove/add amino acids capable of disulfide bonding, to increase its bio-longevity, to alter its secretory characteristics, etc.
  • the antibody fragment must possess a bioactive property, such as binding activity, regulation of binding at the binding domain, etc.
  • Functional or active regions of the antibody may be identified by mutagenesis of a specific region of the protein, followed by expression and testing of the expressed polypeptide. Such methods are readily apparent to a skilled practitioner in the art and can include site-specific mutagenesis of the nucleic acid encoding the antibody fragment.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import” variable domain. Humanization can be essentially performed by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized” antibodies are chimeric antibodies ( US 4,816,567 ), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • Transgenic animals e.g., mice
  • mice that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production
  • homozygous deletion of the antibody heavy chain joining region gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production.
  • Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge.
  • Human antibodies can also be produced in phage display libraries.
  • Antibodies of the invention are preferably administered to a subject in a pharmaceutically acceptable carrier.
  • a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • the pharmaceutically-acceptable carrier include saline, Ringer's solution and dextrose solution.
  • the pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5.
  • Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of antibody being administered.
  • the antibodies can be administered to the subject, patient, or cell by injection (e.g., intravenous, intraperitoneal, subcutaneous, intramuscular), or by other methods such as infusion that ensure its delivery to the bloodstream in an effective form.
  • the antibodies may also be administered by intratumoral or peritumoral routes, to exert local as well as systemic therapeutic effects. Local or intravenous injection is preferred.
  • Effective dosages and schedules for administering the antibodies may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of antibodies that must be administered will vary depending on, for example, the subject that will receive the antibody, the route of administration, the particular type of antibody used and other drugs being administered. A typical daily dosage of the antibody used alone might range from about 1 ( ⁇ g/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above.
  • the efficacy of the therapeutic antibody can be assessed in various ways well known to the skilled practitioner. For instance, the size, number, and/or distribution of cancer in a subject receiving treatment may be monitored using standard tumor imaging techniques.
  • the T-cell receptor can be linked to toxins, drugs, cytokines (see, for example, US 2013/0115191 ), domains recruiting effector cells such as an anti-CD3 domain, etc., in order to execute particular functions on target cells. Moreover, it could be expressed in T cells used for adoptive transfer. Further information can be found in WO 2004/033685A1 and WO 2004/074322A1 . A combination of sTCRs is described in WO 2012/056407A1 . Further methods for the production are disclosed in WO 2013/057586A1 .
  • the peptides and/or the TCRs or antibodies or other binding molecules of the present invention can be used to verify a pathologist's diagnosis of a cancer based on a biopsied sample.
  • the antibodies or TCRs may also be used for in vivo diagnostic assays.
  • the antibody is labeled with a radionucleotide (such as 111 In, 99 Tc, 14 C, 131 I, 3 H, 32 P or 35 S) so that the tumor can be localized using immunoscintiography.
  • a radionucleotide such as 111 In, 99 Tc, 14 C, 131 I, 3 H, 32 P or 35 S
  • antibodies or fragments thereof bind to the extracellular domains of two or more targets of a protein selected from the group consisting of the above-mentioned proteins, and the affinity value (Kd) is less than 1 x 10 ⁇ M.
  • the disease tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin.
  • the fixed or embedded section contains the sample are contacted with a labeled primary antibody and secondary antibody, wherein the antibody is used to detect the expression of the proteins in situ.
  • the mammalian cell lacks or has a reduced level or function of the TAP peptide transporter.
  • Suitable cells that lack the TAP peptide transporter include T2, RMA-S and Drosophila cells.
  • TAP is the transporter associated with antigen processing.
  • the human peptide loading deficient cell line T2 is available from the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852, USA under Catalogue No CRL 1992; the Drosophila cell line Schneider line 2 is available from the ATCC under Catalogue No CRL 19863; the mouse RMA-S cell line is described in Ljunggren et al. (Ljunggren and Karre, 1985).
  • the host cell expresses substantially no MHC class I molecules. It is also preferred that the stimulator cell expresses a molecule important for providing a co-stimulatory signal for T-cells such as any of B7.1, B7.2, ICAM-1 and LFA 3.
  • a molecule important for providing a co-stimulatory signal for T-cells such as any of B7.1, B7.2, ICAM-1 and LFA 3.
  • the nucleic acid sequences of numerous MHC class I molecules and of the co-stimulator molecules are publicly available from the GenBank and EMBL databases.
  • the T cells are CD8-positive T cells.
  • an antigen-presenting cell is transfected to express such an epitope, preferably the cell comprises an expression vector expressing a peptide containing SEQ ID NO: 9.
  • a number of other methods may be used for generating T cells in vitro.
  • autologous tumor-infiltrating lymphocytes can be used in the generation of CTL.
  • Plebanski et al. (Plebanski et al., 1995) made use of autologous peripheral blood lymphocytes (PLBs) in the preparation of T cells.
  • PLBs peripheral blood lymphocytes
  • the production of autologous T cells by pulsing dendritic cells with peptide or polypeptide, or via infection with recombinant virus is possible.
  • B cells can be used in the production of autologous T cells.
  • macrophages pulsed with peptide or polypeptide, or infected with recombinant virus may be used in the preparation of autologous T cells. S.
  • aAPCs artificial antigen presenting cells
  • aAPCs were generated by the coupling of preformed MHC:peptide complexes to the surface of polystyrene particles (microbeads) by biotin:streptavidin biochemistry. This system permits the exact control of the MHC density on aAPCs, which allows to selectively elicit high- or low-avidity antigen-specific T cell responses with high efficiency from blood samples.
  • aAPCs should carry other proteins with co-stimulatory activity like anti-CD28 antibodies coupled to their surface. Furthermore, such aAPC-based systems often require the addition of appropriate soluble factors, e. g. cytokines, like interleukin-12.
  • Allogeneic cells may also be used in the preparation of T cells and a method is described in detail in WO 97/26328 .
  • other cells may be used to present antigens such as CHO cells, baculovirus-infected insect cells, bacteria, yeast, vaccinia-infected target cells.
  • plant viruses may be used (see, for example, Porta et al. (Porta et al., 1994) which describes the development of cowpea mosaic virus as a high-yielding system for the presentation of foreign peptides.
  • the activated T cells that are directed against the peptide for use of the invention are useful in therapy.
  • a further aspect of the invention provides activated T cells obtainable by the foregoing methods of the invention.
  • Activated T cells which are produced by the above method, will selectively recognize a cell that aberrantly expresses a polypeptide that comprises an amino acid sequence of SEQ ID NO: 9.
  • the T cell recognizes the cell by interacting through its TCR with the HLA/peptide-complex (for example, binding).
  • the T cells are useful in a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention wherein the patient is administered an effective number of the activated T cells.
  • the T cells that are administered to the patient may be derived from the patient and activated as described above (i.e. they are autologous T cells). Alternatively, the T cells are not from the patient but are from another individual. Of course, it is preferred if the individual is a healthy individual.
  • healthy individual the inventors mean that the individual is generally in good health, preferably has a competent immune system and, more preferably, is not suffering from any disease that can be readily tested for, and detected.
  • the target cells for the CD8-positive T cells according to the present invention can be cells of the tumor (which sometimes express MHC class II) and/or stromal cells surrounding the tumor (tumor cells) (which sometimes also express MHC class II; (Dengjel et al., 2006)).
  • the T cells of the present invention may be used as active ingredients of a therapeutic composition.
  • a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention comprising administering to the patient an effective number of T cells as defined above.
  • the inventors also mean that the polypeptide is over-expressed compared to levels of expression in normal (healthy) tissues or that the gene is silent in the tissue from which the tumor is derived but in the tumor it is expressed.
  • over-expressed the inventors mean that the polypeptide is present at a level at least 1.2-fold of that present in normal tissue; preferably at least 2-fold, and more preferably at least 5-fold or 10-fold the level present in normal tissue.
  • T cells may be obtained by methods known in the art, e.g. those described above.
  • Another aspect of the present invention includes the use of the peptide for use complexed with MHC to generate a T-cell receptor whose nucleic acid is cloned and is introduced into a host cell, preferably a T cell. This engineered T cell can then be transferred to a patient for therapy of cancer.
  • any molecule of the invention i.e. the peptide for use, nucleic acid, antibody, expression vector, cell, activated T cell, T-cell receptor or the nucleic acid encoding it, is useful for the treatment of disorders, characterized by cells escaping an immune response. Therefore, any molecule of the present invention may be used as medicament or in the manufacture of a medicament. The molecule may be used by itself or combined with other molecule(s) of the invention or (a) known molecule(s).
  • kit comprising:
  • the kit may further comprise one or more of (iii) a buffer, (iv) a diluent, (v) a filter, (vi) a needle, or (v) a syringe.
  • the container is preferably a bottle, a vial, a syringe or test tube; and it may be a multi-use container.
  • the pharmaceutical composition is preferably lyophilized.
  • Kits as disclosed preferably comprise a lyophilized formulation of the present invention in a suitable container and instructions for its reconstitution and/or use.
  • Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the kit and/or container contain/s instructions on or associated with the container that indicates directions for reconstitution and/or use.
  • the label may indicate that the lyophilized formulation is to be reconstituted to peptide concentrations as described above.
  • the label may further indicate that the formulation is useful or intended for subcutaneous administration.
  • the container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation.
  • the kit may further comprise a second container comprising a suitable diluent (e.g., sodium bicarbonate solution).
  • Kits as disclosed may have a single container that contains the formulation of the pharmaceutical compositions for use according to the present invention with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have distinct container for each component.
  • kits as disclosed include a formulation of the invention packaged for use in combination with the co-administration of a second compound (such as adjuvants (e.g. GM-CSF), a chemotherapeutic agent, a natural product, a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof.
  • a second compound such as adjuvants (e.g. GM-CSF), a chemotherapeutic agent, a natural product, a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof.
  • the components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient.
  • the components of the kit may be provided in one or more liquid solutions, preferably, an aqueous solution, more preferably, a sterile aqueous solution.
  • the container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid.
  • the kit will contain a second vial or other container, which allows for separate dosing.
  • the kit may also contain another container for a pharmaceutically acceptable liquid.
  • a therapeutic kit will contain an apparatus (e.g., one or more needles, syringes, eye droppers, pipette, etc.), which enables administration of the agents as disclosed that are components of the present kit.
  • the present formulation is one that is suitable for administration of the peptides by any acceptable route such as oral (enteral), nasal, ophthal, subcutaneous, intradermal, intramuscular, intravenous or transdermal.
  • the administration is s.c., and most preferably i.d. administration may be by infusion pump.
  • the medicament of the invention is preferably used to treat esophageal cancer.
  • the vaccine preferably is a liquid formulation consisting of the individual peptides dissolved in between 20-40% DMSO, preferably about 30-35% DMSO, such as about 33% DMSO.
  • Each peptide to be included into a product is dissolved in DMSO.
  • the concentration of the single peptide solutions has to be chosen depending on the number of peptides to be included into the product.
  • the single peptide-DMSO solutions are mixed in equal parts to achieve a solution containing all peptides to be included in the product with a concentration of -2.5 mg/ml per peptide.
  • the mixed solution is then diluted 1:3 with water for injection to achieve a concentration of 0.826 mg/ml per peptide in 33% DMSO.
  • the diluted solution is filtered through a 0.22 ⁇ m sterile filter. The final bulk solution is obtained.
  • HLA peptide pools from shock-frozen tissue samples were obtained by immune precipitation from solid tissues according to a slightly modified protocol (Falk et al., 1991; Seeger et al., 1999) using the HLA-A*02-specific antibody BB7.2, the HLA-A, -B, - C-specific antibody W6/32, CNBr-activated sepharose, acid treatment, and ultrafiltration.
  • HLA peptide pools as obtained were separated according to their hydrophobicity by reversed-phase chromatography (nanoAcquity UPLC system, Waters) and the eluting peptides were analyzed in LTQ- velos and fusion hybrid mass spectrometers (ThermoElectron) equipped with an ESI source.
  • Peptide pools were loaded directly onto the analytical fused-silica micro-capillary column (75 ⁇ m i.d. x 250 mm) packed with 1.7 ⁇ m C18 reversed-phase material (Waters) applying a flow rate of 400 nL per minute.
  • the peptides were separated using a two-step 180 minute-binary gradient from 10% to 33% B at a flow rate of 300 nL per minute.
  • the gradient was composed of Solvent A (0.1% formic acid in water) and solvent B (0.1% formic acid in acetonitrile).
  • a gold coated glass capillary (PicoTip, New Objective) was used for introduction into the nanoESI source.
  • the LTQ-Orbitrap mass spectrometers were operated in the data-dependent mode using a TOP5 strategy.
  • Label-free relative LC-MS quantitation was performed by ion counting i.e. by extraction and analysis of LC-MS features (Mueller et al., 2007). The method assumes that the peptide's LC-MS signal area correlates with its abundance in the sample. Extracted features were further processed by charge state deconvolution and retention time alignment (Mueller et al., 2008; Sturm et al., 2008). Finally, all LC-MS features were cross-referenced with the sequence identification results to combine quantitative data of different samples and tissues to peptide presentation profiles. The quantitative data were normalized in a two-tier fashion according to central tendency to account for variation within technical and biological replicates.
  • each identified peptide can be associated with quantitative data allowing relative quantification between samples and tissues.
  • all quantitative data acquired for peptide candidates was inspected manually to assure data consistency and to verify the accuracy of the automated analysis.
  • a presentation profile was calculated showing the mean sample presentation as well as replicate variations. The profiles juxtapose esophageal cancer samples to a baseline of normal tissue samples.
  • Presentation profiles of exemplary over-presented peptides are shown in Figure 1 .
  • Presentation scores for exemplary peptides are shown in Table 8.
  • Table 8 Presentation scores.
  • the table lists peptides that are very highly over-presented on tumors compared to a panel of normal tissues (+++), highly over-presented on tumors compared to a panel of normal tissues (++) or over-presented on tumors compared to a panel of normal tissues (+).
  • the panel of normal tissues consisted of: adipose tissue, adrenal gland, artery, vein, bone marrow, brain, central and peripheral nerve, colon, rectum, small intestine incl.
  • duodenum duodenum, esophagus, gallbladder, heart, kidney, liver, lung, lymph node, mononuclear white blood cells, pancreas, peritoneum, pituitary, pleura, salivary gland, skeletal muscle, skin, spleen, stomach, thymus, thyroid gland, trachea, ureter, urinary bladder.
  • RNA from healthy human tissues for RNASeq experiments was obtained from: Asterand (Detroit, USA and Royston, Herts, UK); ProteoGenex Inc. (Culver City, CA, USA); Geneticist Inc. (Glendale, CA, USA); Istituto Nazionale Tumori "Pascale", Molecular Biology and Viral Oncology Unit (IRCCS) (Naples, Italy); University Hospital of Heidelberg (Germany); BioCat GmbH (Heidelberg, Germany).
  • Exemplary expression profiles of source genes as disclosed that are highly over-expressed or exclusively expressed in esophageal cancer are shown in Figures 2 .
  • Expression scores for further exemplary genes are shown in Table 9.
  • Table 9 Expression scores. The table lists peptides from genes that are very highly over-expressed in tumors compared to a panel of normal tissues (+++), highly over-expressed in tumors compared to a panel of normal tissues (++) or over-expressed in tumors compared to a panel of normal tissues (+).
  • the inventors performed investigations using an in vitro T-cell priming assay based on repeated stimulations of CD8+ T cells with artificial antigen presenting cells (aAPCs) loaded with peptide/MHC complexes and anti-CD28 antibody. This way the inventors could show immunogenicity for HLA-A*0201 restricted TUMAPs of the invention, demonstrating that these peptides are T-cell epitopes against which CD8+ precursor T cells exist in humans (Table 10).
  • aAPCs artificial antigen presenting cells
  • the inventors In order to perform in vitro stimulations by artificial antigen presenting cells loaded with peptide-MHC complex (pMHC) and anti-CD28 antibody, the inventors first isolated CD8+ T cells from fresh HLA-A*02 leukapheresis products via positive selection using CD8 microbeads (Miltenyi Biotec, Bergisch-Gladbach, Germany) of healthy donors obtained from the University clinics Mannheim, Germany, after informed consent.
  • CD8 microbeads Miltenyi Biotec, Bergisch-Gladbach, Germany
  • TCM T-cell medium
  • PBMCs and isolated CD8+ lymphocytes were incubated in T-cell medium (TCM) until use consisting of RPMI-Glutamax (Invitrogen, Düsseldorf, Germany) supplemented with 10% heat inactivated human AB serum (PAN-Biotech, Aidenbach, Germany), 100 U/ml Penicillin/100 ⁇ g/ml Streptomycin (Cambrex, Cologne, Germany), 1 mM sodium pyruvate (CC Pro, Oberdorla, Germany), 20 ⁇ g/ml Gentamycin (Cambrex). 2.5 ng/ml IL-7 (PromoCell, Heidelberg, Germany) and 10 U/ml IL-2 (Novartis Pharma, Rhein, Germany) were also added to the TCM at this step.
  • TCM T-cell medium
  • HLA class I peptides For tested HLA class I peptides, in vitro immunogenicity could be demonstrated by generation of peptide specific T-cell lines. Exemplary flow cytometry results after TUMAP-specific multimer staining for two peptides (SEQ ID No 97 and SEQ ID No 101) as disclosed are shown in Figure 3 together with corresponding negative controls. Results for five peptides from the invention are summarized in Table 10A. Table 10A: in vitro immunogenicity of HLA class I peptides as disclosed Exemplary results of in vitro immunogenicity experiments conducted by the applicant for the peptides as disclosed.
  • peptides were synthesized using standard and well-established solid phase peptide synthesis using the Fmoc-strategy. Identity and purity of each individual peptide have been determined by mass spectrometry and analytical RP-HPLC. The peptides were obtained as white to off-white lyophilizates (trifluoro acetate salt) in purities of >50%. All TUMAPs are preferably administered as trifluoro-acetate salts or acetate salts, other salt-forms are also possible.
  • 96 well MAXISorp plates (NUNC) were coated over night with 2ug/ml streptavidin in PBS at room temperature, washed 4x and blocked for 1h at 37°C in 2% BSA containing blocking buffer. Refolded HLA-A*02:01/MLA-001 monomers served as standards, covering the range of 15-500 ng/ml. Peptide-MHC monomers of the UV-exchange reaction were diluted 100 fold in blocking buffer. Samples were incubated for 1h at 37°C, washed four times, incubated with 2ug/ml HRP conjugated anti- ⁇ 2m for 1h at 37°C, washed again and detected with TMB solution that is stopped with NH 2 SO 4 .
  • Candidate peptides that show a high exchange yield are generally preferred for a generation and production of antibodies or fragments thereof, and/or T cell receptors or fragments thereof, as they show sufficient avidity to the MHC molecules and prevent dissociation of the MHC complexes.
  • Table 11 MHC class I binding scores.
  • binders such as antibodies and/or TCRs
  • selection criteria include but are not restricted to exclusiveness of presentation and the density of peptide presented on the cell surface.
  • the quantitation of TUMAP copies per cell in solid tumor samples requires the absolute quantitation of the isolated TUMAP, the efficiency of TUMAP isolation, and the cell count of the tissue sample analyzed.
  • the internal standard is a double-isotope-labelled variant of each peptide, i.e. two isotope-labelled amino acids were included in TUMAP synthesis. It differs from the tumor-associated peptide only in its mass but shows no difference in other physicochemical properties (Anderson et al., 2012).
  • the internal standard was spiked to each MS sample and all MS signals were normalized to the MS signal of the internal standard to level out potential technical variances between MS experiments.
  • the calibration curves were prepared in at least three different matrices, i.e. HLA peptide eluates from natural samples similar to the routine MS samples, and each preparation was measured in duplicate MS runs. For evaluation, MS signals were normalized to the signal of the internal standard and a calibration curve was calculated by logistic regression.
  • the respective samples were also spiked with the internal standard; the MS signals were normalized to the internal standard and quantified using the peptide calibration curve.
  • TUMAP isolation As for any protein purification process, the isolation of proteins from tissue samples is associated with a certain loss of the protein of interest.
  • peptide/MHC complexes were generated for all TUMAPs selected for absolute quantitation.
  • single-isotope-labelled versions of the TUMAPs were used, i.e. one isotope-labelled amino acid was included in TUMAP synthesis.
  • These complexes were spiked into the freshly prepared tissue lysates, i.e. at the earliest possible point of the TUMAP isolation procedure, and then captured like the natural peptide/MHC complexes in the following affinity purification. Measuring the recovery of the single-labelled TUMAPs therefore allows conclusions regarding the efficiency of isolation of individual natural TUMAPs.
  • the efficiency of isolation was analyzed in a low number of samples and was comparable among these tissue samples. In contrast, the isolation efficiency differs between individual peptides. This suggests that the isolation efficiency, although determined in only a limited number of tissue samples, may be extrapolated to any other tissue preparation. However, it is necessary to analyze each TUMAP individually as the isolation efficiency may not be extrapolated from one peptide to others.
  • the inventors applied DNA content analysis. This method is applicable to a wide range of samples of different origin and, most importantly, frozen samples (Alcoser et al., 2011; Forsey and Chaudhuri, 2009; Silva et al., 2013).
  • a tissue sample is processed to a homogenous lysate, from which a small lysate aliquot is taken. The aliquot is divided in three parts, from which DNA is isolated (QiaAmp DNA Mini Kit, Qiagen, Hilden, Germany).
  • the total DNA content from each DNA isolation is quantified using a fluorescence-based DNA quantitation assay (Qubit dsDNA HS Assay Kit, Life Technologies, Darmstadt, Germany) in at least two replicates.
  • a DNA standard curve from aliquots of single healthy blood cells has been generated.
  • the standard curve is used to calculate the total cell content from the total DNA content from each DNA isolation.
  • the mean total cell count of the tissue sample used for peptide isolation is extrapolated considering the known volume of the lysate aliquots and the total lysate volume.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Hospice & Palliative Care (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • General Physics & Mathematics (AREA)

Description

  • The present invention relates to peptides, proteins, nucleic acids and cells for use in immunotherapeutic methods. In particular, the present invention relates to the immunotherapy of cancer. The present invention furthermore relates to a tumor-associated T-cell peptide epitope, alone or in combination with other tumor-associated peptides that can for example serve as active pharmaceutical ingredients of vaccine compositions that stimulate anti-tumor immune responses, or to stimulate T cells ex vivo and transfer into patients. Peptides bound to molecules of the major histocompatibility complex (MHC), or peptides as such, can also be targets of antibodies, soluble T-cell receptors, and other binding molecules.
  • The present invention relates to a peptide sequence derived from HLA class I molecules of human tumor cells that can be used in vaccine compositions for eliciting anti-tumor immune responses, or as target for the development of pharmaceutically/immunologically active compounds and cells.
  • BACKGROUND OF THE INVENTION
  • Esophageal cancer is the eighth most common cancer worldwide, with a five-year prevalence of 464,063 patients in 2012. Mortality rates are very similar to incidence rates (400,169 versus 455,784 in 2012), pointing out the high fatality of esophageal cancer (World Cancer Report, 2014; Ferlay et al., 2013; Bray et al., 2013).
  • Squamous cell carcinoma and adenocarcinoma represent the two most common subtypes of esophageal cancer. Both subtypes are more common in men than in women, but they display distinct geographical distributions. Squamous cell carcinoma is more prevalent in low-resource regions with particularly high incidence rates in the Islamic Republic of Iran, parts of China and Zimbabwe. Adenocarcinoma is the most common type of esophageal cancer among Caucasians and populations with a high socioeconomic status, with the United Kingdom, Australia, the Netherlands and the USA leading the way. The strongest risk factors for the development of esophageal squamous cell carcinoma include alcohol and tobacco consumption, whereas esophageal adenocarcinoma is mainly associated with obesity and gastro-esophageal reflux disease. Incidence rates of esophageal adenocarcinoma are steadily rising in high-income countries, which might be attributed to increasing rates of obesity and gastro-esophageal reflux disease as well as to changes in the classification of tumors at the gastro-esophageal junction. Neuroendocrine carcinoma, adenoid cystic carcinoma, adenosquamous carcinoma, muco-epidermoid carcinoma, mixed adenoneuroendocrine carcinoma, different sarcomas and melanoma represent rarer subtypes of esophageal cancer (World Cancer Report, 2014).
  • The primary treatment strategy for esophageal cancer depends on tumor stage and location, histological type and the medical condition of the patient. Surgery alone is not sufficient, except in a small subgroup of patients with squamous cell carcinoma. In general, surgery should be combined with pre- and eventually post-operative chemotherapy or pre-operative chemoradiation, while pre- or post-operative radiation alone was shown to confer no survival benefit. Chemotherapeutic regimens include oxaliplatin plus fluorouracil, carboplatin plus paclitaxel, cisplatin plus fluorouracil, FOLFOX and cisplatin plus irinotecan. Patients with HER2-positive tumors should be treated according to the guidelines for gastric cancer using a combination of cisplatin, fluorouracil and trastuzumab, as randomized data for targeted therapies in esophageal cancer are very limited (Stahl et al., 2013; Leitlinie Magenkarzinom, 2012).
  • In general, most types of esophageal cancer are well manageable, if patients present with early-stage tumors, whereas therapeutic success is very limited in later stages. Thus, development of new screening protocols could be very effective in reducing esophageal cancer-related mortality rates (World Cancer Report, 2014).
  • Immunotherapy might be a promising novel approach to treat advanced esophageal cancer. Several cancer-associated genes and cancer-testis antigens were shown to be over-expressed in esophageal cancer, including different MAGE genes, NY-ESO-1 and EpCAM (Kimura et al., 2007; Liang et al., 2005b; Inoue et al., 1995; Bujas et al., 2011; Tanaka et al., 1997; Quillien et al., 1997). Those genes represent very interesting targets for immunotherapy and most of them are under investigation for the treatment of other malignancies (ClinicalTrials.gov, 2015). Furthermore, up-regulation of PD-L1 and PD-L2 was described in esophageal cancer, which correlated with poorer prognosis. Thus, esophageal cancer patients with PD-L1-positive tumors might benefit from anti-PD-L1 immunotherapy (Ohigashi et al., 2005).
  • Clinical data on immunotherapeutic approaches in esophageal cancer are still relatively scarce at present, as only a very limited number of early phase clinical trials have been completed (Toomey et al., 2013). A vaccine consisting of three peptides derived from three different cancer-testis antigens (TTK protein kinase, lymphocyte antigen 6 complex locus K and insulin-like growth factor (IGF)-II mRNA binding protein 3) was administered to patients with advanced esophageal cancer in a phase I trial with moderate results (Kono et al., 2009). Intra-tumoral injection of activated T cells after in vitro challenge with autologous malignant cells and interleukin 2 elicited complete or partial tumor responses in four of eleven patients in a phase I/II study (Toh et al., 2000; Toh et al., 2002). Further clinical trials are currently performed to evaluate the impact of different immunotherapies on esophageal cancer, including adoptive cellular therapy (NCT01691625, NCT01691664, NCT01795976, NCT02096614, NCT02457650) vaccination strategies (NCT01143545, NCT01522820) and anti-PD-L1 therapy (NCT02340975) (ClinicalTrials.gov, 2015).
  • Considering the severe side-effects and expense associated with treating cancer, there is a need to identify factors that can be used in the treatment of cancer in general and esophageal cancer in particular. There is also a need to identify factors representing biomarkers for cancer in general and esophageal cancer in particular, leading to better diagnosis of cancer, assessment of prognosis, and prediction of treatment success.
  • Immunotherapy of cancer represents an option of specific targeting of cancer cells while minimizing side effects. Cancer immunotherapy makes use of the existence of tumor associated antigens.
  • The current classification of tumor associated antigens (TAAs) comprises the following major groups:
    1. a) Cancer-testis antigens: The first TAAs ever identified that can be recognized by T cells belong to this class, which was originally called cancer-testis (CT) antigens because of the expression of its members in histologically different human tumors and, among normal tissues, only in spermatocytes/spermatogonia of testis and, occasionally, in placenta. Since the cells of testis do not express class I and II HLA molecules, these antigens cannot be recognized by T cells in normal tissues and can therefore be considered as immunologically tumor-specific. Well-known examples for CT antigens are the MAGE family members and NY-ESO-1.
    2. b) Differentiation antigens: These TAAs are shared between tumors and the normal tissue from which the tumor arose. Most of the known differentiation antigens are found in melanomas and normal melanocytes. Many of these melanocyte lineage-related proteins are involved in biosynthesis of melanin and are therefore not tumor specific but nevertheless are widely used for cancer immunotherapy. Examples include, but are not limited to, tyrosinase and Melan-A/MART-1 for melanoma or PSA for prostate cancer.
    3. c) Over-expressed TAAs: Genes encoding widely expressed TAAs have been detected in histologically different types of tumors as well as in many normal tissues, generally with lower expression levels. It is possible that many of the epitopes processed and potentially presented by normal tissues are below the threshold level for T-cell recognition, while their over-expression in tumor cells can trigger an anticancer response by breaking previously established tolerance. Prominent examples for this class of TAAs are Her-2/neu, survivin, telomerase, or WT1.
    4. d) Tumor-specific antigens: These unique TAAs arise from mutations of normal genes (such as β-catenin, CDK4, etc.). Some of these molecular changes are associated with neoplastic transformation and/or progression. Tumor-specific antigens are generally able to induce strong immune responses without bearing the risk for autoimmune reactions against normal tissues. On the other hand, these TAAs are in most cases only relevant to the exact tumor on which they were identified and are usually not shared between many individual tumors. Tumor-specificity (or -association) of a peptide may also arise if the peptide originates from a tumor- (-associated) exon in case of proteins with tumor-specific (-associated) isoforms.
    5. e) TAAs arising from abnormal post-translational modifications: Such TAAs may arise from proteins which are neither specific nor overexpressed in tumors but nevertheless become tumor associated by posttranslational processes primarily active in tumors. Examples for this class arise from altered glycosylation patterns leading to novel epitopes in tumors as for MUC1 or events like protein splicing during degradation which may or may not be tumor specific.
    6. f) Oncoviral proteins: These TAAs are viral proteins that may play a critical role in the oncogenic process and, because they are foreign (not of human origin), they can evoke a T-cell response. Examples of such proteins are the human papilloma type 16 virus proteins, E6 and E7, which are expressed in cervical carcinoma.
  • T-cell based immunotherapy targets peptide epitopes derived from tumor-associated or tumor-specific proteins, which are presented by molecules of the major histocompatibility complex (MHC). The antigens that are recognized by the tumor specific T lymphocytes, that is, the epitopes thereof, can be molecules derived from all protein classes, such as enzymes, receptors, transcription factors, etc. which are expressed and, as compared to unaltered cells of the same origin, usually up-regulated in cells of the respective tumor.
  • There are two classes of MHC-molecules, MHC class I and MHC class II. MHC class I molecules are composed of an alpha heavy chain and beta-2-microglobulin, MHC class II molecules of an alpha and a beta chain. Their three-dimensional conformation results in a binding groove, which is used for non-covalent interaction with peptides.
    MHC class I molecules can be found on most nucleated cells. They present peptides that result from proteolytic cleavage of predominantly endogenous proteins, defective ribosomal products (DRIPs) and larger peptides. However, peptides derived from endosomal compartments or exogenous sources are also frequently found on MHC class I molecules. This non-classical way of class I presentation is referred to as cross-presentation in the literature (Brossart and Bevan, 1997; Rock et al., 1990). MHC class II molecules can be found predominantly on professional antigen presenting cells (APCs), and primarily present peptides of exogenous or transmembrane proteins that are taken up by APCs e.g. during endocytosis, and are subsequently processed.
    Complexes of peptide and MHC class I are recognized by CD8-positive T cells bearing the appropriate T-cell receptor (TCR), whereas complexes of peptide and MHC class II molecules are recognized by CD4-positive-helper-T cells bearing the appropriate TCR. It is well known that the TCR, the peptide and the MHC are thereby present in a stoichiometric amount of 1:1:1.
  • CD4-positive helper T cells play an important role in inducing and sustaining effective responses by CD8-positive cytotoxic T cells. The identification of CD4-positive T-cell epitopes derived from tumor associated antigens (TAA) is of great importance for the development of pharmaceutical products for triggering anti-tumor immune responses (Gnjatic et al., 2003). At the tumor site, T helper cells, support a cytotoxic T cell- (CTL-) friendly cytokine milieu (Mortara et al., 2006) and attract effector cells, e.g. CTLs, natural killer (NK) cells, macrophages, and granulocytes (Hwang et al., 2007).
  • In the absence of inflammation, expression of MHC class II molecules is mainly restricted to cells of the immune system, especially professional antigen-presenting cells (APC), e.g., monocytes, monocyte-derived cells, macrophages, dendritic cells. In cancer patients, cells of the tumor have been found to express MHC class II molecules (Dengjel et al., 2006).
  • Elongated (longer) peptides of the invention can act as MHC class II active epitopes.
  • T-helper cells, activated by MHC class II epitopes, play an important role in orchestrating the effector function of CTLs in anti-tumor immunity. T-helper cell epitopes that trigger a T-helper cell response of the TH1 type support effector functions of CD8-positive killer T cells, which include cytotoxic functions directed against tumor cells displaying tumor-associated peptide/MHC complexes on their cell surfaces. In this way tumor-associated T-helper cell peptide epitopes, alone or in combination with other tumor-associated peptides, can serve as active pharmaceutical ingredients of vaccine compositions that stimulate anti-tumor immune responses.
  • It was shown in mammalian animal models, e.g., mice, that even in the absence of CD8-positive T lymphocytes, CD4-positive T cells are sufficient for inhibiting manifestation of tumors via inhibition of angiogenesis by secretion of interferon-gamma (IFNγ) (Beatty and Paterson, 2001; Mumberg et al., 1999). There is evidence for CD4 T cells as direct anti-tumor effectors (Braumuller et al., 2013; Tran et al., 2014).
  • Since the constitutive expression of HLA class II molecules is usually limited to immune cells, the possibility of isolating class II peptides directly from primary tumors was previously not considered possible. However, Dengjel et al. were successful in identifying a number of MHC Class II epitopes directly from tumors ( WO 2007/028574 , EP 1 760 088 B1 ).
  • Since both types of response, CD8 and CD4 dependent, contribute jointly and synergistically to the anti-tumor effect, the identification and characterization of tumor-associated antigens recognized by either CD8+ T cells (ligand: MHC class I molecule + peptide epitope) or by CD4-positive T-helper cells (ligand: MHC class II molecule + peptide epitope) is important in the development of tumor vaccines.
  • For an MHC class I peptide to trigger (elicit) a cellular immune response, it also must bind to an MHC-molecule. This process is dependent on the allele of the MHC-molecule and specific polymorphisms of the amino acid sequence of the peptide. MHC-class-I-binding peptides are usually 8-12 amino acid residues in length and usually contain two conserved residues ("anchors") in their sequence that interact with the corresponding binding groove of the MHC-molecule. In this way each MHC allele has a "binding motif" determining which peptides can bind specifically to the binding groove.
  • In the MHC class I dependent immune reaction, peptides not only have to be able to bind to certain MHC class I molecules expressed by tumor cells, they subsequently also have to be recognized by T cells bearing specific T cell receptors (TCR).
  • For proteins to be recognized by T-lymphocytes as tumor-specific or -associated antigens, and to be used in a therapy, particular prerequisites must be fulfilled. The antigen should be expressed mainly by tumor cells and not, or in comparably small amounts, by normal healthy tissues. In a preferred embodiment, the peptide should be over-presented by tumor cells as compared to normal healthy tissues. It is furthermore desirable that the respective antigen is not only present in a type of tumor, but also in high concentrations (i.e. copy numbers of the respective peptide per cell). Tumor-specific and tumor-associated antigens are often derived from proteins directly involved in transformation of a normal cell to a tumor cell due to their function, e.g. in cell cycle control or suppression of apoptosis. Additionally, downstream targets of the proteins directly causative for a transformation may be up-regulated und thus may be indirectly tumor-associated. Such indirect tumor-associated antigens may also be targets of a vaccination approach (Singh-Jasuja et al., 2004). It is essential that epitopes are present in the amino acid sequence of the antigen, in order to ensure that such a peptide ("immunogenic peptide"), being derived from a tumor associated antigen, leads to an in vitro or in vivo T-cell-response.
  • Basically, any peptide able to bind an MHC molecule may function as a T-cell epitope. A prerequisite for the induction of an in vitro or in vivo T-cell-response is the presence of a T cell having a corresponding TCR and the absence of immunological tolerance for this particular epitope.
  • Therefore, TAAs are a starting point for the development of a T cell based therapy including but not limited to tumor vaccines. The methods for identifying and characterizing the TAAs are usually based on the use of T-cells that can be isolated from patients or healthy subjects, or they are based on the generation of differential transcription profiles or differential peptide expression patterns between tumors and normal tissues. However, the identification of genes over-expressed in tumor tissues or human tumor cell lines, or selectively expressed in such tissues or cell lines, does not provide precise information as to the use of the antigens being transcribed from these genes in an immune therapy. This is because only an individual subpopulation of epitopes of these antigens are suitable for such an application since a T cell with a corresponding TCR has to be present and the immunological tolerance for this particular epitope needs to be absent or minimal. In a very preferred embodiment of the invention it is therefore important to select only those over- or selectively presented peptides against which a functional and/or a proliferating T cell can be found. Such a functional T cell is defined as a T cell, which upon stimulation with a specific antigen can be clonally expanded and is able to execute effector functions ("effector T cell").
  • In case of targeting peptide-MHC by specific TCRs (e.g. soluble TCRs) and antibodies according to the invention, the immunogenicity of the underlying peptides is secondary. In these cases, the presentation is the determining factor.
  • Rizzolo et al. (in: Conventional and microwave-assisted SPPS approach: a comparative synthesis of PTHrP(1-34)NH2D3 J Pept Sci. 2011 Oct;17(10):708-14) relates to a synthesis method for polypeptides, and compares Fmoc/tBu MW-assisted SPPS of 1-34 N-terminal fragment of parathyroid hormone-related peptide (PTHrP) with its conventional SPPS carried out at RT. During the stepwise elongation of the resin-bound peptide, monitoring was conducted by performing MW-assisted mini-cleavages and analyzing them by UPLC-ESI-MS. Identification of some deletion sequences was helpful to recognize critical couplings and as such helped to guide the introduction of MW irradiations to these stages. Only peptide fragments during synthesis are disclosed. Nothing is stated with respect to the use of the peptide as claimed in medicine.
  • US 2005/033023 discloses a 10mer peptide FLHHLIAEIH (PTR-2, SEQ ID NO: 3) overlapping with SEQ ID NO: 9 of the present application, HLIAEIHTA. In US 2005/033023 , PTH-rP molecules were screened with the "Parker" algorithm (BIMAS) in order to predict peptides having high theoretical HLA-A(*)02.01 binding motifs. PTR-2 was found to have the highest HLA-A(*)02.01 binding affinity, nevertheless, the motif of the present peptide was not identified in US 2005/033023 .
  • SUMMARY OF THE INVENTION
  • In a first aspect of the present invention, the present invention relates to a peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  • The following tables show the peptide for use according to the present invention, respective SEQ ID NOs, and the prospective source (underlying) genes for these peptides. All peptides in Table 1 and Table 2 bind to HLA-A*02. The peptides in Table 2 have been disclosed before in large listings as results of high-throughput screenings with high error rates or calculated using algorithms, but have not been associated with cancer at all before. The peptides in Table 3 are additional peptides that may be useful in combination with the other peptides for use of the invention. The peptides in Table 4 are furthermore useful in the diagnosis and/or treatment of various other malignancies that involve an over-expression or over-presentation of the respective underlying polypeptide. Table 1: Peptides as disclosed, SEQ ID NO: 9 is for use according to the invention
    SEQ ID No Sequence Gene ID(s) Official Gene Symbol(s)
    1 STYGGGLSV 3861, 3868 KRT14, KRT16
    2 SLYNLGGSKRISI 3852 KRT5
    3 TASAITPSV 3852 KRT5
    4 ALFGTILEL 2769 GNA15
    5 NLMASQPQL 5317 PKP1
    6 LLSGDLIFL 2709 GJB5
    7 SIFEGLLSGV 2709 GJB5
    8 ALLDGGSEAYWRV 84985 FAM83A
    9 HLIAEIHTA 5744 PTHLH
    10 SLDENSDQQV 6273 S100A2
    11 ALWLPTDSATV 3914 LAMB3
    12 GLASRILDA 3914 LAMB3
    13 SLSPVILGV 26525 IL36RN
    14 RLPNAGTQV 3655 ITGA6
    15 LLANGVYAA 55107 ANO1
    16 VLAEGGEGV 10630 PDPN
    17 MISRTPEV 2155, 28396, 3500, 3501, 3502, 3503, 3507 F7, IGHV4-31, IGHG1, IGHG2, IGHG3, IGHG4, IGHM
    3507 IGHM
    18 FLLDQVQLGL 83882 TSPAN10
    19 GLAPFLLNAV 101060689, 154761, 285966 FAM115C
    20 IIEVDPDTKEML 100505503, 402057, 442216, 6218 RPS17L, RPS17P16, RPS17P5, RPS17
    21 IVREFLTAL 27297 CRCP
    22 KLNDTYVNV 23306 TMEM194A
    23 KLSDSATYL No associated gene
    24 LLFAGTMTV 29785 CYP2S1
    25 LLPPPPPPA 9509 ADAMTS2
    26 MLAEKLLQA 2195 FAT1
    27 NLREGDQLL 113146 AHNAK2
    28 SLDGFTIQV 4939 OAS2
    29 SLDGTELQL 284114 TMEM102
    30 SLNGNQVTV 79832 QSER1
    31 VLPKLYVKL 100996747, 441502, 6231, 643003, 644928, 728937, 729188 RPS26P11, RPS26, RPS26P28, RPS26P15, RPS26P25, RPS26P58
    32 YMLDIFHEV 3038 HAS3
    33 GLDVTSLRPFDL 2316 FLNA
    34 SLVSEQLEPA 11187 PKP3
    35 LLRFSQDNA 51056 LAP3
    36 FLLRFSQDNA 51056 LAP3
    37 YTQPFSHYGQAL 6051 RNPEP
    38 IAAIRGFLV 83451 ABHD11
    39 LVRDTQSGSL 871 SERPINH1
    40 GLAFSL YQA 871 SERPINH1
    41 GLESEELEPEEL 8106 PABPN1
    42 TQTAVITRI 81610 FAM83D
    43 KVVGKDYLL 832 CAPZB
    44 ATGNDRKEAAENS L 7531 YWHAE
    45 MLTELEKAL 6279 S100A8
    46 YTAQIGADIAL 64499, 7177 TPSB2, TPSAB1
    47 VLASGFLTV 79183 TTPAL
    48 SMHQMLDQTL 7168 TPM1
    49 GLMKDIVGA 8942 KYNU
    50 GMNPHQTPAQL 471 ATIC
    51 KLFGHL TSA 57157 PHTF2
    52 VAIGGVDGNVRL 9948 WDR1
    53 VWTGLTLV 396 ARHGDIA
    54 YQDLLNVKM 1674, 4741, 4744, 4747, 7431, 9118 DES, NEFM, NEFH, NEFL, VIM, INA
    55 GAIDLLHNV 115362 GBP5
    56 ALVEVTEHV 54972 TMEM132A
    57 GLAPNTPGKA 9055 PRC1
    58 LILESIPVV 5597 MAPK6
    59 SLLDTLREV 9989 PPP4R1
    60 VVMEELLKV 23191 CYFIP1
    61 TQTTH ELTI 5093 PCBP1
    62 ALYEYQPLQI 4331 MNAT1
    63 LAYTLGVKQL 158078, 1915, 1917 EEF1A1P5, EEF1A1, EEF1A2
    64 GLTDVIRDV 80028 FBXL18
    65 YWGGFLYQRL 4074 M6PR
    66 LLDEKVQSV 57616 TSHZ3
    67 SMNGGVFAV 23657 SLC7A11
    68 PAVLQSSGLYSL 28396, 3500, 3501, 3502, 3503, 3507 IGHV4-31, IGHG1, IGHG2, IGHG3, IGHG4, IGHM
    69 GLL VGSEKVTM 3861, 3868, 644945 KRT14, KRT16, KRT16P3
    70 FVLDTSESV 1291, 1292 COL6A1, COL6A2
    71 ASDPILYRPVAV 5315 PKM
    72 FLPPAQVTV 65083 NOL6
    73 KITEAIQYV 6095 RORA
    74 ILASLATSV 10844 TUBGCP2
    75 GLMDDVDFKA 10525 HYOU1
    76 KVADYIPQL 2744 GLS
    Table 2: Additional peptides as disclosed with no prior known cancer association
    SEQ ID No Sequence Gene ID(s) Official Gene Symbol(s)
    77 VLVPYEPPQV 8626 TP63
    78 KVANIIAEV 5910 RAP1GDS1
    79 GQDVGRYQV 6748 SSR4
    80 ALQEALENA 9631 NUP155
    81 AVLPHVDQV 23379 KIAA0947
    82 HLLGHLEQA 63977 PRDM15
    83 ALADGVVSQA 27238 GPKOW
    84 SLAESLDQA 22894 DIS3
    85 NIIELVHQV 6850 SYK
    86 GLL TEIRAV 9263 STK17A
    87 FLDNGPKTI 1982 EIF4G2
    88 GLWEQENHL 79768 KATNBL1
    89 SLADSLYNL 23271 CAMSAP2
    90 SIYEYYHAL 3091 HIF1A
    91 KLIDDVHRL 6734 SRPR
    92 SILRHVAEV 1965 EIF2S1
    93 VLINTSVTL 23036 ZNF292
    Table 3: Peptides useful for e.g. personalized cancer therapies
    SEQ ID No Sequence Gene ID(s) Official Gene Symbol(s)
    94 TLLQEQGTKTV 286887, 3852, 3853, 3854 KRT6C, KRT5, KRT6A, KRT6B
    95 LIQDRVAEV 3914 LAMB3
    96 GAAVRIGSVL 9150 CTDP1
    97 ELDRTPPEV 23450 SF3B3
    98 VLFPNLKTV 646 BNC1
    99 RVAPEEHPVL 440915, 60, 641455, 71,728378 POTEKP, ACTB, POTEM, ACTG1, POTEF
    100 GLYPDAFAPV 1991 ELANE
    101 AMTQLLAGV 3371 TNC
  • The present invention furthermore generally relates to the peptides according to the present invention for use in the treatment of proliferative diseases, such as, for example, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • Disclosed are the peptides - alone or in combination - from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 93. More preferred are the peptides - alone or in combination - selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 76 (see Table 1), and their uses in the immunotherapy of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia, and preferably esophageal cancer.
  • Disclosed are the peptides - alone or in combination - selected from the group consisting of SEQ ID No. 1, 2, 3, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 16, 17, 18, 19, 25, 26, 30, 32, 34, 37, 40, 51, 55, 57, 58, 59, 62, 81, and 82, and their uses in the immunotherapy of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia, and preferably esophageal cancer. Further particularly preferred is the peptide for use according to SEQ ID NO: 9.
  • As shown in the following Table 4A, many of the peptides as disclosed are also found on other tumor types and can, thus, also be used in the immunotherapy of other indications. Also refer to Figure 1 and Example 1. Table 4A: Peptides as disclosed and their specific uses in other proliferative diseases, especially in other cancerous diseases. SEQ ID NO: 9 is for use according to the invention. The table shows for selected peptides on which additional tumor types they were found and either over-presented on more than 5% of the measured tumor samples, or presented on more than 5% of the measured tumor samples with a ratio of geometric means tumor vs normal tissues being larger than 3. Over-presentation is defined as higher presentation on the tumor sample as compared to the normal sample with highest presentation. Normal tissues against which over-presentation was tested were: adipose tissue, adrenal gland, artery, bone marrow, brain, central nerve, colon, duodenum, esophagus, gallbladder, heart, kidney, liver, lung, lymph node, mononuclear white blood cells, pancreas, peripheral nerve, peritoneum, pituitary, pleura, rectum, salivary gland, skeletal muscle, skin, small intestine, spleen, stomach, thymus, thyroid gland, trachea, ureter, urinary bladder, vein.
    SEQ ID No. Sequence Other relevant organs / diseases
    2 SLYNLGGSKRISI NSCLC
    3 TASAITPSV NSCLC, Urinary bladder cancer
    4 ALFGTILEL NSCLC
    7 SIFEGLLSGV Urinary bladder cancer
    8 ALLDGGSEAYWRV NSCLC, OC
    9 HLIAEIHTA NSCLC
    11 ALWLPTDSATV NSCLC, Melanoma
    12 GLASRILDA Urinary bladder cancer
    13 SLSPVILGV Uterine Cancer
    15 LLANGVYAA HCC
    17 MISRTPEV NSCLC, RCC, HCC
    22 KLNDTYVNV SCLC, Brain Cancer, HCC
    26 MLAEKLLQA CRC
    29 SLDGTELQL BRCA
    31 VLPKLYVKL GC
    32 YMLDIFHEV Urinary bladder cancer
    33 GLDVTSLRPFDL GC
    34 SLVSEQLEPA CRC, Urinary bladder cancer
    35 LLRFSQDNA GC, HCC
    36 FLLRFSQDNA GC
    37 YTQPFSHYGQAL GC, PC
    38 IAAIRGFLV GC
    39 LVRDTQSGSL GC
    40 GLAFSL YQA NSCLC, PC, BRCA, Urinary bladder cancer
    41 GLESEELEPEEL GC
    42 TQTAVITRI GC
    45 MLTELEKAL GC
    46 YTAQIGADIAL GC
    47 VLASGFLTV Urinary bladder cancer
    49 GLMKDIVGA HCC
    50 GMNPHQTPAQL GC
    51 KLFGHL TSA Gallbladder Cancer, Bile Duct Cancer
    52 VAIGGVDGNVRL GC
    54 YQDLLNVKM RCC, GC
    55 GAIDLLHNV GC
    56 ALVEVTEHV BRCA
    57 GLAPNTPGKA NSCLC, SCLC, BRCA, Melanoma
    58 LILESIPW NSCLC, Melanoma
    61 TQTTH ELTI SCLC, Leukemia
    62 ALYEYQPLQI NSCLC, HCC, Urinary bladder cancer
    63 LAYTLGVKQL GC
    66 LLDEKVQSV Brain Cancer, Melanoma
    67 SMNGGVFAV Brain Cancer, HCC
    68 PAVLQSSGLYSL GC, PC
    69 GLL VGSEKVTM PC
    70 FVLDTSESV GC, HCC, Melanoma, OC
    71 ASDPILYRPVAV GC, PC
    72 FLPPAQVTV NSCLC, GC, HCC, Leukemia, Melanoma
    73 KITEAIQYV BRCA
    74 ILASLATSV CRC, HCC, Urinary bladder cancer
    75 GLMDDVDFKA BRCA, Melanoma, Urinary bladder cancer
    76 KVADYIPQL NSCLC, SCLC
    77 VLVPYEPPQV NSCLC, Urinary bladder cancer
    78 KVANIIAEV PC, Leukemia, OC
    80 ALQEALENA NSCLC, SCLC, Brain Cancer, CRC, HCC, Leukemia, BRCA, OC
    81 AVLPHVDQV Brain Cancer
    82 HLLGHLEQA NSCLC, HCC, Leukemia, BRCA
    83 ALADGVVSQA Brain Cancer, GC, Melanoma, Urinary bladder cancer
    85 NIIELVHQV Leukemia
    86 GLL TEIRAV Brain Cancer, Urinary bladder cancer
    87 FLDNGPKTI Brain Cancer, PC, OC
    88 GLWEQENHL NSCLC, BRCA
    89 SLADSLYNL Brain Cancer, BRCA, Urinary bladder cancer, Gallbladder Cancer, Bile Duct Cancer
    91 KLIDDVHRL PC, PrC
    92 SILRHVAEV NSCLC, CRC, HCC, Gallbladder Cancer, Bile Duct Cancer
    94 TLLQEQGTKTV NSCLC
    NSCLC= non-small cell lung cancer, SCLC= small cell lung cancer, RCC= kidney cancer, CRC= colon or rectum cancer, GC= stomach cancer, HCC= liver cancer, PC= pancreatic cancer, PrC= prostate cancer, leukemia, BrCa=breast cancer
    Table 4B: Peptides as disclosed and their specific uses in other proliferative diseases, especially in other cancerous diseases (amendment of Table 4). The table shows, like Table 4A, for selected peptides on which additional tumor types they were found showing over-presentation (including specific presentation) on more than 5% of the measured tumor samples, or presentation on more than 5% of the measured tumor samples with a ratio of geometric means tumor vs normal tissues being larger than 3. Over-presentation is defined as higher presentation on the tumor sample as compared to the normal sample with highest presentation. Normal tissues against which over-presentation was tested were: adipose tissue, adrenal gland, artery, bone marrow, brain, central nerve, colon, duodenum, esophagus, eye, gallbladder, heart, kidney, liver, lung, lymph node, mononuclear white blood cells, pancreas, parathyroid gland, peripheral nerve, peritoneum, pituitary, pleura, rectum, salivary gland, skeletal muscle, skin, small intestine, spleen, stomach, thyroid gland, trachea, ureter, urinary bladder, vein.
    SEQ ID No Sequence Additional Entities
    1 STYGGGLSV NSCLC, Melanoma, HNSCC
    2 SLYNLGGSKRISI Urinary bladder cancer, HNSCC
    3 TASAITPSV HNSCC
    4 ALFGTILEL Urinary bladder cancer, Gallbladder Cancer, Bile Duct Cancer, AML, HNSCC
    5 NLMASQPQL HNSCC
    6 LLSGDLIFL HNSCC
    7 SIFEGLLSGV NSCLC, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    8 ALLDGGSEAYWR V Urinary bladder cancer, HNSCC
    10 SLDENSDQQV Urinary bladder cancer, HNSCC
    11 ALWLPTDSATV Gallbladder Cancer, Bile Duct Cancer
    13 SLSPVILGV NSCLC, Melanoma, Urinary bladder cancer, HNSCC
    14 RLPNAGTQV Melanoma
    15 LLANGVYAA Urinary bladder cancer, Uterine Cancer, Gallbladder Cancer, Bile Duct Cancer
    16 VLAEGGEGV Brain Cancer, Melanoma, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    17 MISRTPEV Urinary bladder cancer
    18 FLLDQVQLGL Melanoma, NHL, HNSCC
    19 GLAPFLLNAV Melanoma, NHL, HNSCC
    20 IIEVDPDTKEML HNSCC
    22 KLNDTYVNV BRCA
    23 KLSDSATYL Melanoma
    25 LLPPPPPPA Gallbladder Cancer, Bile Duct Cancer, NHL, HNSCC
    28 SLDGFTIQV BRCA, Melanoma, AML
    29 SLDGTELQL Uterine Cancer, NHL
    30 SLNGNQVTV BRCA, Melanoma, Urinary bladder cancer, Uterine Cancer, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    32 YMLDIFHEV Gallbladder Cancer, Bile Duct Cancer, HNSCC
    34 SLVSEQLEPA HNSCC
    40 GLAFSL YQA CRC, Melanoma, Uterine Cancer, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    42 TQTAVITRI HNSCC
    48 SMHQMLDQTL GC
    51 KLFGHL TSA Gallbladder Cancer, Bile Duct Cancer
    53 VVVTGLTLV GC, Urinary bladder cancer
    55 GAIDLLHNV SCLC, Melanoma, NHL
    56 ALVEVTEHV RCC, Uterine Cancer, Gallbladder Cancer, Bile Duct Cancer
    57 GLAPNTPGKA Urinary bladder cancer, Uterine Cancer, HNSCC
    58 LILESIPVV SCLC, CLL, Urinary bladder cancer, Uterine Cancer, NHL, HNSCC
    59 SLLDTLREV HNSCC
    62 ALYEYQPLQI SCLC, BRCA, Melanoma, OC, Gallbladder Cancer, Bile Duct Cancer, NHL
    66 LLDEKVQSV Urinary bladder cancer, HNSCC
    67 SMNGGVFAV NSCLC, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    68 PAVLQSSGLYSL NHL
    69 GLL VGSEKVTM HNSCC
    72 FLPPAQVTV CLL, Urinary bladder cancer, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    74 ILASLATSV BRCA, HNSCC
    75 GLMDDVDFKA GC, HCC, Gallbladder Cancer, Bile Duct Cancer, NHL, HNSCC
    76 KVADYIPQL RCC, BRCA, Melanoma, Gallbladder Cancer, Bile Duct Cancer, NHL
    77 VLVPYEPPQV NHL, HNSCC
    78 KVANIIAEV Urinary bladder cancer, HNSCC
    79 GQDVGRYQV SCLC, PC, PrC, CLL, BRCA, OC, Urinary bladder cancer, AML, NHL
    80 ALQEALENA Melanoma, Uterine Cancer, Gallbladder Cancer, Bile Duct Cancer, HNSCC
    81 AVLPHVDQV Uterine Cancer, NHL
    82 HLLGHLEQA RCC
    83 ALADGVVSQA Uterine Cancer
    84 SLAESLDQA Melanoma, Uterine Cancer, AML, NHL, HNSCC
    85 NIIELVHQV CLL
    86 GLL TEIRAV Melanoma, Gallbladder Cancer, Bile Duct Cancer, AML, NHL, HNSCC
    87 FLDNGPKTI Urinary bladder cancer, HNSCC
    88 GLWEQENHL CRC, Uterine Cancer, AML, HNSCC
    89 SLADSLYNL Melanoma
    90 SIYEYYHAL NHL, HNSCC
    92 SILRHVAEV BRCA, Melanoma, AML, NHL
    NSCLC= non-small cell lung cancer, SCLC= small cell lung cancer, RCC= kidney cancer, CRC= colon or rectum cancer, GC= stomach cancer, HCC= liver cancer, PC= pancreatic cancer, PrC= prostate cancer, BRCA=breast cancer, OC= ovarian cancer, NHL= non-Hodgkin lymphoma, AML= acute myeloid leukemia, CLL= chronic lymphocytic leukemia, HNSCC= head and neck squamous cell carcinoma.
  • Thus, another aspect of the present invention relates to the use of at least one peptide as disclosed according to any one of SEQ ID No. 1, 2, 3, 4, 7, 8, 9, 11, 13, 17, 40, 57, 58, 62, 67, 72, 76, 77, 80, 82, 88, 92 and 94 for the - in one preferred embodiment combined - treatment of non-small cell lung cancer.
  • Thus, another aspect of the present invention relates to the use of the peptides according to the present invention for the - preferably combined - treatment of a proliferative disease selected from the group of esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • The present invention furthermore relates to a peptide for use according to the present invention that has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I.
  • The present invention relates to peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  • The present invention further relates to the peptide for use according_to the present invention, wherein said peptide includes non-peptide bonds.
  • The present invention further relates to the peptide for use according to the present invention, wherein said peptide is part of a fusion protein comprising the N-terminal 80 amino acids of the HLA-DR antigen-associated invariant chain (li), or fused to (or into the sequence of) an antibody, such as, for example, an antibody that is specific for dendritic cells.
  • The present invention further relates to a nucleic acid, encoding the peptide for use according to the present invention. The present invention further relates to the nucleic acid according to the present invention that is DNA, cDNA, PNA, RNA or combinations thereof.
  • The present invention further relates to an expression vector expressing a nucleic acid according to the present invention.
  • The present invention further relates to a peptide according to the present invention, a nucleic acid according to the present invention or an expression vector according to the present invention for use in the treatment of diseases and in medicine, in particular in the treatment of cancer.
  • The present invention further relates to antibodies that are specific against the peptide for use according to the present invention or complexes of said peptide for use according to the present invention with MHC, and methods of making these.
  • The present invention further relates to T-cell receptors (TCRs), in particular soluble TCR (sTCRs) and cloned TCRs engineered into autologous or allogeneic T cells, and methods of making these, as well as NK cells or other cells bearing said TCR or cross-reacting with said TCRs.
  • The antibodies and TCRs are additional embodiments of the immunotherapeutic use of the peptides according to the invention at hand.
  • The present invention further relates to a host cell comprising a nucleic acid according to the present invention or an expression vector as described before. The present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably is a dendritic cell.
  • The present invention further relates to a method for producing a peptide for use according to the present invention, said method comprising culturing the host cell according to the present invention, and isolating the peptide from said host cell or its culture medium.
  • The present invention further relates to said method according to the present invention, wherein the antigen is loaded onto class I MHC molecules expressed on the surface of a suitable antigen-presenting cell or artificial antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell.
  • The present invention further relates to the method according to the present invention, wherein the antigen-presenting cell comprises an expression vector expressing said peptide containing SEQ ID No. 9.
  • The present invention further relates to activated T cells, produced by the method according to the present invention, wherein said T cell selectively recognizes a cell which expresses a polypeptide comprising an amino acid sequence for use according to the present invention.
  • Disclosed is a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising any amino acid sequence according to the present invention, the method comprising administering to the patient an effective number of T cells as produced according to the present invention.
  • The present invention further relates to the peptide as described, the nucleic acid according to the present invention, the expression vector according to the present invention, the cell according to the present invention, the activated T lymphocyte, the T cell receptor or the antibody or other peptide- and/or peptide-MHC-binding molecules according to the present invention for use as a medicament or in the manufacture of a medicament. Preferably, said medicament is active against cancer.
  • Preferably, said medicament is a cellular therapy, a vaccine or a protein based on a soluble TCR or antibody.
  • The present invention further relates to a use according to the present invention, wherein said cancer cells are esophageal cancer, lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia, and preferably esophageal cancer cells.
  • For example, an antibody or soluble TCR can be used to stain sections of the tumor to detect the presence of a peptide of interest in complex with MHC.
  • Optionally, the antibody carries a further effector function such as an immune stimulating domain or toxin.
  • Distinct polymorphisms in PTHLH were shown to be associated with lung cancer risk and prognosis (Manenti et al., 2000). Up-regulation of PTHLH in a C57BL/6-mouse-derived model of spontaneously metastatic mammary cancer was described as potentially being involved in metastatic dissemination of breast cancer (Johnstone et al., 2015). PTHLH was shown to be up-regulated in oral squamous cell carcinoma, chondroid neoplasms, adult T-cell leukemia/lymphoma and clear cell renal cell carcinomas (Bellon et al., 2013; Yang et al., 2013a; Yao et al., 2014; Lv et al., 2014). PTHLH up-regulation was shown to be associated with poor pathologic differentiation and poor prognosis in patients with head and neck squamous cell carcinoma (Lv et al., 2014). PTHLH was shown to be up-regulated through p38 MAPK signaling, which contributes to colon cancer cell extravasation of the lung by caspase-independent death in endothelial cells of the lung microsvasculature (Urosevic et al., 2014). PTHLH was shown to be significantly differentially expressed in squamous cell carcinoma compared with normal skin (Prasad et al., 2014). PTHLH was described as a part of a four-gene signature associated with survival among patients with early-stage non-small cell lung cancer (Chang et al., 2012). Disruption of anti-proliferative function by frameshift mutations of PTHLH was described to contribute to the development of early colorectal cancer in patients with hereditary non-polyposis colorectal cancer (Yamaguchi et al., 2006). PTHLH up-regulation was shown to be associated with poor outcome both in overall survival and disease-free survival for clear cell renal cell carcinoma patients who underwent nephrectomy (Yao et al., 2014). PTHLH was shown to positively modulate cell cycle progression and to change the expression of proteins involved in cell cycle regulation via ERK1/2, p38, MAPK, and PI3K signaling pathways in the colorectal adenocarcinoma cell line Caco-2 (Calvo et al., 2014).
  • DETAILED DESCRIPTION OF THE INVENTION
  • Stimulation of an immune response is dependent upon the presence of antigens recognized as foreign by the host immune system. The discovery of the existence of tumor associated antigens has raised the possibility of using a host's immune system to intervene in tumor growth. Various mechanisms of harnessing both the humoral and cellular arms of the immune system are currently being explored for cancer immunotherapy.
  • Specific elements of the cellular immune response are capable of specifically recognizing and destroying tumor cells. The isolation of T-cells from tumor-infiltrating cell populations or from peripheral blood suggests that such cells play an important role in natural immune defense against cancer. CD8-positive T-cells in particular, which recognize class I molecules of the major histocompatibility complex (MHC)-bearing peptides of usually 8 to 10 amino acid residues derived from proteins or defect ribosomal products (DRIPS) located in the cytosol, play an important role in this response. The MHC-molecules of the human are also designated as human leukocyte-antigens (HLA).
  • As used herein and except as noted otherwise all terms are defined as given below.
  • The term "T-cell response" means the specific proliferation and activation of effector functions induced by a peptide in vitro or in vivo. For MHC class I restricted cytotoxic T cells, effector functions may be lysis of peptide-pulsed, peptide-precursor pulsed or naturally peptide-presenting target cells, secretion of cytokines, preferably Interferon-gamma, TNF-alpha, or IL-2 induced by peptide, secretion of effector molecules, preferably granzymes or perforins induced by peptide, or degranulation.
  • The term "peptide" is used herein to designate a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The peptides are preferably 9 amino acids in length, but can be as short as 8 amino acids in length, and as long as 10, 11, 12, 13, or 14 or longer, and in case of MHC class II peptides (elongated variants of the peptides as disclosed) they can be as long as 15, 16, 17, 18, 19 or 20 or more amino acids in length.
  • Furthermore, the term "peptide" shall include salts of a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. Preferably, the salts are pharmaceutical acceptable salts of the peptides, such as, for example, the chloride or acetate (trifluoroacetate) salts. It has to be noted that the salts of the peptides according to the present invention differ substantially from the peptides in their state(s) in vivo, as the peptides are not salts in vivo.
  • The term "peptide" shall also include "oligopeptide". The term "oligopeptide" is used herein to designate a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The length of the oligopeptide is not critical to the invention, as long as the correct epitope or epitopes are maintained therein. The oligopeptides are typically less than about 30 amino acid residues in length, and greater than about 15 amino acids in length.
  • The term "polypeptide" designates a series of amino acid residues, connected one to the other typically by peptide bonds between the alpha-amino and carbonyl groups of the adjacent amino acids. The length of the polypeptide is not critical to the invention as long as the correct epitopes are maintained. In contrast to the terms peptide or oligopeptide, the term polypeptide is meant to refer to molecules containing more than about 30 amino acid residues.
  • A peptide, oligopeptide, protein or polynucleotide coding for such a molecule is "immunogenic" (and thus is an "immunogen" within the present invention), if it is capable of inducing an immune response. In the case of the present invention, immunogenicity is more specifically defined as the ability to induce a T-cell response. Thus, an "immunogen" would be a molecule that is capable of inducing an immune response, and in the case of the present invention, a molecule capable of inducing a T-cell response. In another aspect, the immunogen can be the peptide, the complex of the peptide with MHC, oligopeptide, and/or protein that is used to raise specific antibodies or TCRs against it.
  • A class I T cell "epitope" requires a short peptide that is bound to a class I MHC receptor, forming a ternary complex (MHC class I alpha chain, beta-2-microglobulin, and peptide) that can be recognized by a T cell bearing a matching T-cell receptor binding to the MHC/peptide complex with appropriate affinity. Peptides binding to MHC class I molecules are typically 8-14 amino acids in length, and most typically 9 amino acids in length.
  • In humans there are three different genetic loci that encode MHC class I molecules (the MHC-molecules of the human are also designated human leukocyte antigens (HLA)): HLA-A, HLA-B, and HLA-C. HLA-A*01, HLA-A*02, and HLA-B*07 are examples of different MHC class I alleles that can be expressed from these loci. Table 5: Expression frequencies F of HLA-A*02 and HLA-A*24 and the most frequent HLA-DR serotypes. Frequencies are deduced from haplotype frequencies Gf within the American population adapted from Mori et al. (Mori et al., 1997) employing the Hardy-Weinberg formula F = 1 - (1-Gf)2. Combinations of A*02 or A*24 with certain HLA-DR alleles might be enriched or less frequent than expected from their single frequencies due to linkage disequilibrium. For details refer to Chanock et al. (Chanock et al., 2004).
    Allele Population Calculated phenotype from allele frequency
    A*02 Caucasian (North America) 49.1%
    A*02 African American (North America) 34.1%
    A*02 Asian American (North America) 43.2%
    A*02 Latin American (North American) 48.3%
    DR1 Caucasian (North America) 19.4%
    DR2 Caucasian (North America) 28.2%
    DR3 Caucasian (North America) 20.6%
    DR4 Caucasian (North America) 30.7%
    DR5 Caucasian (North America) 23.3%
    DR6 Caucasian (North America) 26.7%
    DR7 Caucasian (North America) 24.8%
    DR8 Caucasian (North America) 5.7%
    DR9 Caucasian (North America) 2.1%
    DR1 African (North) American 13.20%
    DR2 African (North) American 29.80%
    DR3 African (North) American 24.80%
    DR4 African (North) American 11.10%
    DR5 African (North) American 31.10%
    DR6 African (North) American 33.70%
    DR7 African (North) American 19.20%
    DR8 African (North) American 12.10%
    DR9 African (North) American 5.80%
    DR1 Asian (North) American 6.80%
    DR2 Asian (North) American 33.80%
    DR3 Asian (North) American 9.20%
    DR4 Asian (North) American 28.60%
    DR5 Asian (North) American 30.00%
    DR6 Asian (North) American 25.10%
    DR7 Asian (North) American 13.40%
    DR8 Asian (North) American 12.70%
    DR9 Asian (North) American 18.60%
    DR1 Latin (North) American 15.30%
    DR2 Latin (North) American 21.20%
    DR3 Latin (North) American 15.20%
    DR4 Latin (North) American 36.80%
    DR5 Latin (North) American 20.00%
    DR6 Latin (North) American 31.10%
    DR7 Latin (North) American 20.20%
    DR8 Latin (North) American 18.60%
    DR9 Latin (North) American 2.10%
    A*24 Philippines 65%
    A*24 Russia Nenets 61%
    A*24:02 Japan 59%
    A*24 Malaysia 58%
    A*24:02 Philippines 54%
    A*24 India 47%
    A*24 South Korea 40%
    A*24 Sri Lanka 37%
    A*24 China 32%
    A*24:02 India 29%
    A*24 Australia West 22%
    A*24 USA 22%
    A*24 Russia Samara 20%
    A*24 South America 20%
    A*24 Europe 18%
  • The peptide for use of the invention, preferably when included into a vaccine of the invention as described herein bind to A*02. A vaccine may also include pan-binding MHC class II peptides. Therefore, the vaccine of the invention can be used to treat cancer in patients that are A*02 positive, whereas no selection for MHC class II allotypes is necessary due to the pan-binding nature of these peptides.
  • If A*02 peptides of the invention are combined with peptides binding to another allele, for example A*24, a higher percentage of any patient population can be treated compared with addressing either MHC class I allele alone. While in most populations less than 50% of patients could be addressed by either allele alone, a vaccine comprising HLA-A*24 and HLA-A*02 epitopes can treat at least 60% of patients in any relevant population. Specifically, the following percentages of patients will be positive for at least one of these alleles in various regions: USA 61%, Western Europe 62%, China 75%, South Korea 77%, Japan 86% (calculated from www.allelefrequencies.net).
  • In a preferred embodiment, the term "nucleotide sequence" refers to a heteropolymer of deoxyribonucleotides.
  • The nucleotide sequence coding for a particular peptide, oligopeptide, or polypeptide may be naturally occurring or they may be synthetically constructed. Generally, DNA segments encoding the peptides, polypeptides, and proteins of this invention are assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene that is capable of being expressed in a recombinant transcriptional unit comprising regulatory elements derived from a microbial or viral operon.
  • As used herein the term "a nucleotide coding for (or encoding) a peptide" refers to a nucleotide sequence coding for the peptide including artificial (man-made) start and stop codons compatible for the biological system the sequence is to be expressed by, for example, a dendritic cell or another cell system useful for the production of TCRs.
  • As used herein, reference to a nucleic acid sequence includes both single stranded and double stranded nucleic acid. Thus, for example for DNA, the specific sequence, unless the context indicates otherwise, refers to the single strand DNA of such sequence, the duplex of such sequence with its complement (double stranded DNA) and the complement of such sequence.
  • The term "coding region" refers to that portion of a gene which either naturally or normally codes for the expression product of that gene in its natural genomic environment, i.e., the region coding in vivo for the native expression product of the gene.
  • The coding region can be derived from a non-mutated ("normal"), mutated or altered gene, or can even be derived from a DNA sequence, or gene, wholly synthesized in the laboratory using methods well known to those of skill in the art of DNA synthesis.
  • The term "expression product" means the polypeptide or protein that is the natural translation product of the gene and any nucleic acid sequence coding equivalents resulting from genetic code degeneracy and thus coding for the same amino acid(s).
  • The term "fragment", when referring to a coding sequence, means a portion of DNA comprising less than the complete coding region, whose expression product retains essentially the same biological function or activity as the expression product of the complete coding region.
  • The term "DNA segment" refers to a DNA polymer, in the form of a separate fragment or as a component of a larger DNA construct, which has been derived from DNA isolated at least once in substantially pure form, i.e., free of contaminating endogenous materials and in a quantity or concentration enabling identification, manipulation, and recovery of the segment and its component nucleotide sequences by standard biochemical methods, for example, by using a cloning vector. Such segments are provided in the form of an open reading frame uninterrupted by internal non-translated sequences, or introns, which are typically present in eukaryotic genes. Sequences of non-translated DNA may be present downstream from the open reading frame, where the same do not interfere with manipulation or expression of the coding regions.
  • The term "primer" means a short nucleic acid sequence that can be paired with one strand of DNA and provides a free 3'-OH end at which a DNA polymerase starts synthesis of a deoxyribonucleotide chain.
  • The term "promoter" means a region of DNA involved in binding of RNA polymerase to initiate transcription.
  • The term "isolated" means that the material is removed from its original environment (e.g., the natural environment, if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • The polynucleotides, and recombinant or immunogenic polypeptides, disclosed in accordance with the present invention may also be in "purified" form. The term "purified" does not require absolute purity; rather, it is intended as a relative definition, and can include preparations that are highly purified or preparations that are only partially purified, as those terms are understood by those of skill in the relevant art. For example, individual clones isolated from a cDNA library have been conventionally purified to electrophoretic homogeneity. Purification of starting material or natural material to at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated. Furthermore, a claimed polypeptide which has a purity of preferably 99.999%, or at least 99.99% or 99.9%; and even desirably 99% by weight or greater is expressly encompassed.
  • The nucleic acids and polypeptide expression products disclosed according to the present invention, as well as expression vectors containing such nucleic acids and/or such polypeptides, may be in "enriched form". As used herein, the term "enriched" means that the concentration of the material is at least about 2, 5, 10, 100, or 1000 times its natural concentration (for example), advantageously 0.01%, by weight, preferably at least about 0.1% by weight. Enriched preparations of about 0.5%, 1%, 5%, 10%, and 20% by weight are also contemplated. The sequences, constructs, vectors, clones, and other materials comprising the present invention can advantageously be in enriched or isolated form. The term "active fragment" means a fragment, usually of a peptide, polypeptide or nucleic acid sequence, that generates an immune response (i.e., has immunogenic activity) when administered, alone or optionally with a suitable adjuvant or in a vector, to an animal, such as a mammal, for example, a rabbit or a mouse, and also including a human, such immune response taking the form of stimulating a T-cell response within the recipient animal, such as a human. Alternatively, the "active fragment" may also be used to induce a T-cell response in vitro.
  • As used herein, the terms "portion", "segment" and "fragment", when used in relation to polypeptides, refer to a continuous sequence of residues, such as amino acid residues, which sequence forms a subset of a larger sequence. For example, if a polypeptide were subjected to treatment with any of the common endopeptidases, such as trypsin or chymotrypsin, the oligopeptides resulting from such treatment would represent portions, segments or fragments of the starting polypeptide. When used in relation to polynucleotides, these terms refer to the products produced by treatment of said polynucleotides with any of the endonucleases.
  • In accordance with the present invention, the term "percent identity" or "percent identical", when referring to a sequence, means that a sequence is compared to a claimed or described sequence after alignment of the sequence to be compared (the "Compared Sequence") with the described or claimed sequence (the "Reference Sequence"). The percent identity is then determined according to the following formula: percent identity = 100 1 C / R
    Figure imgb0001
    wherein C is the number of differences between the Reference Sequence and the Compared Sequence over the length of alignment between the Reference Sequence and the Compared Sequence, wherein
    • (i) each base or amino acid in the Reference Sequence that does not have a corresponding aligned base or amino acid in the Compared Sequence and
    • (ii) each gap in the Reference Sequence and
    • (iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, constitutes a difference and
    • (iiii) the alignment has to start at position 1 of the aligned sequences;
    and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid.
  • If an alignment exists between the Compared Sequence and the Reference Sequence for which the percent identity as calculated above is about equal to or greater than a specified minimum Percent Identity, then the Compared Sequence has the specified minimum percent identity to the Reference Sequence even though alignments may exist in which the herein above calculated percent identity is less than the specified percent identity.
  • As mentioned above, the present invention thus provides peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine. The peptide for use of the invention has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I.
  • In the present invention, the term "homologous" refers to the degree of identity (see percent identity above) between sequences of two amino acid sequences, i.e. peptide or polypeptide sequences. The aforementioned "homology" is determined by comparing two sequences aligned under optimal conditions over the sequences to be compared. Such a sequence homology can be calculated by creating an alignment using, for example, the ClustalW algorithm. Commonly available sequence analysis software, more specifically, Vector NTI, GENETYX or other tools are provided by public databases.
  • Of course, the peptide for use according to the present invention will have the ability to bind to a molecule of the human major histocompatibility complex (MHC) class I . Binding of a peptide or a variant to a MHC complex may be tested by methods known in the art.
  • Preferably, when the T cells specific for a peptide for use according to the present invention are tested against the substituted peptides, the peptide concentration at which the substituted peptides achieve half the maximal increase in lysis relative to background is no more than about 1 mM, preferably no more than about 1 µM, more preferably no more than about 1 nM, and still more preferably no more than about 100 pM, and most preferably no more than about 10 pM. It is also preferred that the substituted peptide be recognized by T cells from more than one individual, at least two, and more preferably three individuals.
  • In one embodiment of the present invention, the peptide for use is part of a fusion protein which comprises the 80 N-terminal amino acids of the HLA-DR antigen-associated invariant chain (p33, in the following "li") as derived from the NCBI, GenBank Accession number X00497. In other fusions, the peptides for use of the present invention can be fused to an antibody as described herein, or a functional part thereof, in particular into a sequence of an antibody, so as to be specifically targeted by said antibody, or, for example, to or into an antibody that is specific for dendritic cells as described herein.
  • In addition, the peptide for use may include non-peptide bonds.
  • In a reverse peptide bond amino acid residues are not joined by peptide (-CO-NH-) linkages but the peptide bond is reversed. Such retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al (1997) (Meziere et al., 1997). This approach involves making pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Meziere et al. (Meziere et al., 1997) show that for MHC binding and T helper cell responses, these pseudopeptides are useful. Retro-inverse peptides, which contain NH-CO bonds instead of CO-NH peptide bonds, are much more resistant to proteolysis.
  • A non-peptide bond is, for example, -CH2-NH, -CH2S-, -CH2CH2-, -CH=CH-, -COCH2-, - CH(OH)CH2-, and -CH2SO-. US 4,897,445 provides a method for the solid phase synthesis of non-peptide bonds (-CH2-NH) in polypeptide chains which involves polypeptides synthesized by standard procedures and the non-peptide bond synthesized by reacting an amino aldehyde and an amino acid in the presence of NaCNBH3.
  • Peptides comprising the sequences described above may be synthesized with additional chemical groups present at their amino and/or carboxy termini, to enhance the stability, bioavailability, and/or affinity of the peptides. For example, hydrophobic groups such as carbobenzoxyl, dansyl, or t-butyloxycarbonyl groups may be added to the peptides' amino termini. Likewise, an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the peptides' amino termini. Additionally, the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the peptides' carboxy termini.
  • Further, the peptides as disclosed may be synthesized to alter their steric configuration. For example, the D-isomer of one or more of the amino acid residues of the peptide may be used, rather than the usual L-isomer. Still further, at least one of the amino acid residues of the peptides of the invention may be substituted by one of the well-known non-naturally occurring amino acid residues. Alterations such as these may serve to increase the stability, bioavailability and/or binding action of the peptides of the invention.
  • Generally, peptides (at least those containing peptide linkages between amino acid residues) may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lukas et al. (Lukas et al., 1981) and by references as cited therein. Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide. Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine). Where glutamine or asparagine are C-terminal residues, use is made of the 4,4'-dimethoxybenzhydryl group for protection of the side chain amido functionalities. The solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalizing agent). The peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N, N-dicyclohexylcarbodiimide/1hydroxybenzotriazole mediated coupling procedure. All coupling and deprotection reactions are monitored using ninhydrin, trinitrobenzene sulphonic acid or isotin test procedures. Upon completion of synthesis, peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50 % scavenger mix. Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible (see, for example, (Bruckdorfer et al., 2004), and the references as cited therein).
  • Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide. Any scavengers present are removed by a simple extraction procedure which on lyophilization of the aqueous phase affords the crude peptide free of scavengers. Reagents for peptide synthesis are generally available from e.g. Calbiochem-Novabiochem (Nottingham, UK).
  • Purification may be performed by any one, or a combination of, techniques such as recrystallization, size exclusion chromatography, ion-exchange chromatography, hydrophobic interaction chromatography and (usually) reverse-phase high performance liquid chromatography using e.g. acetonitrile/water gradient separation.
  • Analysis of peptides may be carried out using thin layer chromatography, electrophoresis, in particular capillary electrophoresis, solid phase extraction (CSPE), reverse-phase high performance liquid chromatography, amino-acid analysis after acid hydrolysis and by fast atom bombardment (FAB) mass spectrometric analysis, as well as MALDI and ESI-Q-TOF mass spectrometric analysis.
  • In order to select over-presented peptides, a presentation profile is calculated showing the median sample presentation as well as replicate variation. The profile juxtaposes samples of the tumor entity of interest to a baseline of normal tissue samples. Each of these profiles can then be consolidated into an over-presentation score by calculating the p-value of a Linear Mixed-Effects Model (Pinheiro et al., 2015) adjusting for multiple testing by False Discovery Rate (Benjamini and Hochberg, 1995) (cf. Example 1).
  • For the identification and relative quantitation of HLA ligands by mass spectrometry, HLA molecules from shock-frozen tissue samples were purified and HLA-associated peptides were isolated. The isolated peptides were separated and sequences were identified by online nano-electrospray-ionization (nanoESI) liquid chromatography-mass spectrometry (LC-MS) experiments. The resulting peptide sequences were verified by comparison of the fragmentation pattern of natural tumor-associated peptides (TUMAPs) recorded from esophageal cancer samples (N = 16 A*02-positive samples) with the fragmentation patterns of corresponding synthetic reference peptides of identical sequences. Since the peptides were directly identified as ligands of HLA molecules of primary tumors, these results provide direct evidence for the natural processing and presentation of the identified peptides on primary cancer tissue obtained from 16 esophageal cancer patients.
  • The discovery pipeline XPRESIDENT® v2.1 (see, for example, US 2013-0096016 ) allows the identification and selection of relevant over-presented peptide vaccine candidates based on direct relative quantitation of HLA-restricted peptide levels on cancer tissues in comparison to several different non-cancerous tissues and organs. This was achieved by the development of label-free differential quantitation using the acquired LC-MS data processed by a proprietary data analysis pipeline, combining algorithms for sequence identification, spectral clustering, ion counting, retention time alignment, charge state deconvolution and normalization.
  • Presentation levels including error estimates for each peptide and sample were established. Peptides exclusively presented on tumor tissue and peptides over-presented in tumor versus non-cancerous tissues and organs have been identified.
  • HLA-peptide complexes from esophageal cancer tissue samples were purified and HLA-associated peptides were isolated and analyzed by LC-MS (see examples). All TUMAPs contained in the present application were identified with this approach on primary esophageal cancer samples confirming their presentation on primary esophageal cancer.
  • TUMAPs identified on multiple esophageal cancer and normal tissues were quantified using ion-counting of label-free LC-MS data. The method assumes that LC-MS signal areas of a peptide correlate with its abundance in the sample. All quantitative signals of a peptide in various LC-MS experiments were normalized based on central tendency, averaged per sample and merged into a bar plot, called presentation profile. The presentation profile consolidates different analysis methods like protein database search, spectral clustering, charge state deconvolution (decharging) and retention time alignment and normalization.
  • In addition to over-presentation of the peptide, mRNA expression of the underlying gene was tested. mRNA data were obtained via RNASeq analyses of normal tissues and cancer tissues (see Example 2). An additional source of normal tissue data was a database of publicly available RNA expression data from around 3000 normal tissue samples (Lonsdale, 2013). Peptides which are derived from proteins whose coding mRNA is highly expressed in cancer tissue, but very low or absent in vital normal tissues, were preferably included in the present invention.
  • Furthermore, the discovery pipeline XPRESIDENT® v2. allows for a direct absolute quantitation of MHC-, preferably HLA-restricted, peptide levels on cancer or other infected tissues. Briefly, the total cell count was calculated from the total DNA content of the analyzed tissue sample. The total peptide amount for a TUMAP in a tissue sample was measured by nanoLC-MS/MS as the ratio of the natural TUMAP and a known amount of an isotope-labelled version of the TUMAP, the so-called internal standard. The efficiency of TUMAP isolation was determined by spiking peptide:MHC complexes of all selected TUMAPs into the tissue lysate at the earliest possible point of the TUMAP isolation procedure and their detection by nanoLC-MS/MS following completion of the peptide isolation procedure. The total cell count and the amount of total peptide were calculated from triplicate measurements per tissue sample. The peptide-specific isolation efficiencies were calculated as an average from 10 spike experiments each measured as a triplicate (see Example 6 and Table 12).
  • The present invention provides a peptide for use in treating cancers/tumors, preferably esophageal cancer that over- or exclusively present the peptide for use of the invention. The peptide was shown by mass spectrometry to be naturally presented by HLA molecules on primary human esophageal cancer samples.
  • Many of the source gene/proteins (also designated "full-length proteins" or "underlying proteins") from which the peptides are derived were shown to be highly over-expressed in cancer compared with normal tissues - "normal tissues" in relation to this invention shall mean either healthy esophagus cells or other normal tissue cells, demonstrating a high degree of tumor association of the source genes (see Example 2). Moreover, the peptides themselves are strongly over-presented on tumor tissue - "tumor tissue" in relation to this invention shall mean a sample from a patient suffering from esophageal cancer, but not on normal tissues (see Example 1).
  • HLA-bound peptides can be recognized by the immune system, specifically T lymphocytes. T cells can destroy the cells presenting the recognized HLA/peptide complex, e.g. esophageal cancer cells presenting the derived peptides.
  • The peptide for use of the present invention has been shown to be capable of stimulating T cell responses and/or is over-presented and thus can be used for the production of antibodies and/or TCRs, such as soluble TCRs, according to the present invention (see Example 3, Example 4). Furthermore, the peptides when complexed with the respective MHC can be used for the production of antibodies and/or TCRs, in particular sTCRs, according to the present invention, as well. Respective methods are well known to the person of skill, and can be found in the respective literature as well. Thus, the peptide for use of the present invention is useful for generating an immune response in a patient by which tumor cells can be destroyed. An immune response in a patient can be induced by direct administration of the described peptides or suitable precursor substances (e.g. elongated peptides, proteins, or nucleic acids encoding these peptides) to the patient, ideally in combination with an agent enhancing the immunogenicity (i.e. an adjuvant). The immune response originating from such a therapeutic vaccination can be expected to be highly specific against tumor cells because the target peptides of the present invention are not presented on normal tissues in comparable copy numbers, preventing the risk of undesired autoimmune reactions against normal cells in the patient.
  • The present description further relates to T-cell receptors (TCRs) comprising an alpha chain and a beta chain ("alpha/beta TCRs"). The present description also relates to nucleic acids, vectors and host cells for expressing TCRs and peptides of the present description; and methods of using the same.
  • The term "T-cell receptor" (abbreviated TCR) refers to a heterodimeric molecule comprising an alpha polypeptide chain (alpha chain) and a beta polypeptide chain (beta chain), wherein the heterodimeric receptor is capable of binding to a peptide antigen presented by an HLA molecule. The term also includes so-called gamma/delta TCRs.
  • In one embodiment the description provides a method of producing a TCR as described herein, the method comprising culturing a host cell capable of expressing the TCR under conditions suitable to promote expression of the TCR.
  • The description in another aspect relates to methods according to the description, wherein the antigen is loaded onto class I MHC molecules expressed on the surface of a suitable antigen-presenting cell or artificial antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell or the antigen is loaded onto class I MHC tetramers by tetramerizing the antigen/class I MHC complex monomers.
  • The alpha and beta chains of alpha/beta TCR's, and the gamma and delta chains of gamma/delta TCRs, are generally regarded as each having two "domains", namely variable and constant domains. The variable domain consists of a concatenation of variable region (V), and joining region (J). The variable domain may also include a leader region (L). Beta and delta chains may also include a diversity region (D). The alpha and beta constant domains may also include C-terminal transmembrane (TM) domains that anchor the alpha and beta chains to the cell membrane.
  • With respect to gamma/delta TCRs, the term "TCR gamma variable domain" as used herein refers to the concatenation of the TCR gamma V (TRGV) region without leader region (L), and the TCR gamma J (TRGJ) region, and the term TCR gamma constant domain refers to the extracellular TRGC region, or to a C-terminal truncated TRGC sequence. Likewise the term "TCR delta variable domain" refers to the concatenation of the TCR delta V (TRDV) region without leader region (L) and the TCR delta D/J (TRDD/TRDJ) region, and the term "TCR delta constant domain" refers to the extracellular TRDC region, or to a C-terminal truncated TRDC sequence.
  • TCRs of the present description preferably bind to a peptide-HLA molecule complex with a binding affinity (KD) of about 100 µM or less, about 50 µM or less, about 25 µM or less, or about 10 µM or less. More preferred are high affinity TCRs having binding affinities of about 1 µM or less, about 100 nM or less, about 50 nM or less, about 25 nM or less. Non-limiting examples of preferred binding affinity ranges for TCRs of the present invention include about 1 nM to about 10 nM; about 10 nM to about 20 nM; about 20 nM to about 30 nM; about 30 nM to about 40 nM; about 40 nM to about 50 nM; about 50 nM to about 60 nM; about 60 nM to about 70 nM; about 70 nM to about 80 nM; about 80 nM to about 90 nM; and about 90 nM to about 100 nM.
  • As used herein in connect with TCRs of the present description, "specific binding" and grammatical variants thereof are used to mean a TCR having a binding affinity (KD) for a peptide-HLA molecule complex of 100 µM or less.
  • Alpha/beta heterodimeric TCRs of the present description may have an introduced disulfide bond between their constant domains. Preferred TCRs of this type include those which have a TRAC constant domain sequence and a TRBC1 or TRBC2 constant domain sequence except that Thr 48 of TRAC and Ser 57 of TRBC1 or TRBC2 are replaced by cysteine residues, the said cysteines forming a disulfide bond between the TRAC constant domain sequence and the TRBC1 or TRBC2 constant domain sequence of the TCR.
  • With or without the introduced inter-chain bond mentioned above, alpha/beta heterodimeric TCRs of the present description may have a TRAC constant domain sequence and a TRBC1 or TRBC2 constant domain sequence, and the TRAC constant domain sequence and the TRBC1 or TRBC2 constant domain sequence of the TCR may be linked by the native disulfide bond between Cys4 of exon 2 of TRAC and Cys2 of exon 2 of TRBC1 or TRBC2.
  • TCRs of the present description may comprise a detectable label selected from the group consisting of a radionuclide, a fluorophore and biotin. TCRs of the present description may be conjugated to a therapeutically active agent, such as a radionuclide, a chemotherapeutic agent, or a toxin.
  • In an embodiment, a TCR of the present description having at least one mutation in the alpha chain and/or having at least one mutation in the beta chain has modified glycosylation compared to the non-mutated TCR.
  • In an embodiment, a TCR comprising at least one mutation in the TCR alpha chain and/or TCR beta chain has a binding affinity for, and/or a binding half-life for, a peptide-HLA molecule complex, which is at least double that of a TCR comprising the non-mutated TCR alpha chain and/or non-mutated TCR beta chain. Affinity-enhancement of tumor-specific TCRs, and its exploitation, relies on the existence of a window for optimal TCR affinities. The existence of such a window is based on observations that TCRs specific for HLA-A2-restricted pathogens have KD values that are generally about 10-fold lower when compared to TCRs specific for HLA-A2-restricted tumor-associated self-antigens. It is now known, although tumor antigens have the potential to be immunogenic, because tumors arise from the individual's own cells only mutated proteins or proteins with altered translational processing will be seen as foreign by the immune system. Antigens that are upregulated or overexpressed (so called self-antigens) will not necessarily induce a functional immune response against the tumor: T-cells expressing TCRs that are highly reactive to these antigens will have been negatively selected within the thymus in a process known as central tolerance, meaning that only T-cells with low-affinity TCRs for self-antigens remain.
  • In one aspect, to obtain T-cells expressing TCRs of the present description, nucleic acids encoding TCR-alpha and/or TCR-beta chains of the present description are cloned into expression vectors, such as gamma retrovirus or lentivirus. The recombinant viruses are generated and then tested for functionality, such as antigen specificity and functional avidity. An aliquot of the final product is then used to transduce the target T-cell population (generally purified from patient PBMCs), which is expanded before infusion into the patient.
  • In another aspect, to obtain T-cells expressing TCRs of the present description, TCR RNAs are synthesized by techniques known in the art, e.g., in vitro transcription systems. The in vitro-synthesized TCR RNAs are then introduced into primary CD8+ T-cells obtained from healthy donors by electroporation to re-express tumor specific TCR-alpha and/or TCR-beta chains.
  • To increase the expression, nucleic acids encoding TCRs of the present description may be operably linked to strong promoters, such as retroviral long terminal repeats (LTRs), cytomegalovirus (CMV), murine stem cell virus (MSCV) U3, phosphoglycerate kinase (PGK), β-actin, ubiquitin, and a simian virus 40 (SV40)/CD43 composite promoter, elongation factor (EF)-1a and the spleen focus-forming virus (SFFV) promoter. In a preferred embodiment, the promoter is heterologous to the nucleic acid being expressed.
  • In addition to strong promoters, TCR expression cassettes of the present description may contain additional elements that can enhance transgene expression, including a central polypurine tract (cPPT), which promotes the nuclear translocation of lentiviral constructs (Follenzi et al., 2000), and the woodchuck hepatitis virus posttranscriptional regulatory element (wPRE), which increases the level of transgene expression by increasing RNA stability (Zufferey et al., 1999).
  • The alpha and beta chains of a TCR of the present invention may be encoded by nucleic acids located in separate vectors, or may be encoded by polynucleotides located in the same vector.
  • Achieving high-level TCR surface expression requires that both the TCR-alpha and TCR-beta chains of the introduced TCR be transcribed at high levels. To do so, the TCR-alpha and TCR-beta chains of the present description may be cloned into bi-cistronic constructs in a single vector, which has been shown to be capable of overcoming this obstacle. The use of a viral intra-ribosomal entry site (IRES) between the TCR-alpha and TCR-beta chains results in the coordinated expression of both chains, because the TCR-alpha and TCR-beta chains are generated from a single transcript that is broken into two proteins during translation, ensuring that an equal molar ratio of TCR-alpha and TCR-beta chains are produced. (Schmitt et al. 2009).
  • Nucleic acids encoding TCRs of the present description may be codon optimized to increase expression from a host cell. Redundancy in the genetic code allows some amino acids to be encoded by more than one codon, but certain codons are less "optimal" than others because of the relative availability of matching tRNAs as well as other factors (Gustafsson et al., 2004). Modifying the TCR-alpha and TCR-beta gene sequences such that each amino acid is encoded by the optimal codon for mammalian gene expression, as well as eliminating mRNA instability motifs or cryptic splice sites, has been shown to significantly enhance TCR-alpha and TCR-beta gene expression (Scholten et al., 2006).
  • Furthermore, mispairing between the introduced and endogenous TCR chains may result in the acquisition of specificities that pose a significant risk for autoimmunity. For example, the formation of mixed TCR dimers may reduce the number of CD3 molecules available to form properly paired TCR complexes, and therefore can significantly decrease the functional avidity of the cells expressing the introduced TCR (Kuball et al., 2007).
  • To reduce mispairing, the C-terminus domain of the introduced TCR chains of the present description may be modified in order to promote interchain affinity, while decreasing the ability of the introduced chains to pair with the endogenous TCR. These strategies may include replacing the human TCR-alpha and TCR-beta C-terminus domains with their murine counterparts (murinized C-terminus domain); generating a second interchain disulfide bond in the C-terminus domain by introducing a second cysteine residue into both the TCR-alpha and TCR-beta chains of the introduced TCR (cysteine modification); swapping interacting residues in the TCR-alpha and TCR-beta chain C-terminus domains ("knob-in-hole"); and fusing the variable domains of the TCR-alpha and TCR-beta chains directly to CD3ζ (CD3ζ fusion). (Schmitt et al. 2009).
  • In an embodiment, a host cell is engineered to express a TCR of the present description. In preferred embodiments, the host cell is a human T-cell or T-cell progenitor. In some embodiments the T-cell or T-cell progenitor is obtained from a cancer patient. In other embodiments the T-cell or T-cell progenitor is obtained from a healthy donor. Host cells of the present description can be allogeneic or autologous with respect to a patient to be treated. In one embodiment, the host is a gamma/delta T-cell transformed to express an alpha/beta TCR.
  • A "pharmaceutical composition" is a composition suitable for administration to a human being in a medical setting. Preferably, a pharmaceutical composition is sterile and produced according to GMP guidelines.
  • The pharmaceutical compositions comprise the peptides either in the free form or in the form of a pharmaceutically acceptable salt (see also above). As used herein, "a pharmaceutically acceptable salt" refers to a derivative of the disclosed peptides wherein the peptide is modified by making acid or base salts of the agent. For example, acid salts are prepared from the free base (typically wherein the neutral form of the drug has a neutral -NH2 group) involving reaction with a suitable acid. Suitable acids for preparing acid salts include both organic acids, e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methane sulfonic acid, ethane sulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid phosphoric acid and the like. Conversely, preparation of basic salts of acid moieties which may be present on a peptide are prepared using a pharmaceutically acceptable base such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine or the like.
  • In an especially preferred embodiment, the pharmaceutical compositions comprise the peptides as salts of acetic acid (acetates), trifluoro acetates or hydrochloric acid (chlorides).
  • Preferably, the medicament for use of the present invention is an immunotherapeutics such as a vaccine. It may be administered directly into the patient, into the affected organ or systemically i.d., i.m., s.c., i.p. and i.v., or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation of immune cells derived from the patient, which are then re-administered to the patient. If the nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2. The peptide may be substantially pure, or combined with an immune-stimulating adjuvant (see below) or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, for example liposomes. The peptide may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see WO 95/18145 and (Longenecker et al., 1993)). The peptide may also be tagged, may be a fusion protein, or may be a hybrid molecule. The peptides whose sequence is given in the present invention are expected to stimulate CD4 or CD8 T cells. However, stimulation of CD8 T cells is more efficient in the presence of help provided by CD4 T-helper cells. Thus, for MHC Class I epitopes that stimulate CD8 T cells the fusion partner or sections of a hybrid molecule suitably provide epitopes which stimulate CD4-positive T cells. CD4- and CD8-stimulating epitopes are well known in the art and include those identified in the present invention.
  • In one aspect, the vaccine for use comprises at least one peptide having the amino acid sequence set forth in SEQ ID No. 9, and at least one additional peptide, preferably two to 50, more preferably two to 25, even more preferably two to 20 and most preferably two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen or eighteen peptides. The peptide(s) may be derived from one or more specific TAAs and may bind to MHC class I molecules.
  • A further aspect of the invention provides a nucleic acid (for example a polynucleotide) encoding a peptide for use of the invention. The polynucleotide may be, for example, DNA, cDNA, PNA, RNA or combinations thereof, either single- and/or double-stranded, or native or stabilized forms of polynucleotides, such as, for example, polynucleotides with a phosphorothioate backbone and it may or may not contain introns so long as it codes for the peptide. Of course, only peptides that contain naturally occurring amino acid residues joined by naturally occurring peptide bonds are encodable by a polynucleotide. A still further aspect of the invention provides an expression vector expressing a peptide for use according to the invention.
  • A variety of methods have been developed to link polynucleotides, especially DNA, to vectors for example via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc. New Haven, CN, USA.
  • A desirable method of modifying the DNA encoding the polypeptide of the invention employs the polymerase chain reaction as disclosed by Saiki RK, et al. (Saiki et al., 1988).This method may be used for introducing the DNA into a suitable vector, for example by engineering in suitable restriction sites, or it may be used to modify the DNA in other useful ways as is known in the art. If viral vectors are used, pox- or adenovirus vectors are preferred.
  • The DNA (or in the case of retroviral vectors, RNA) may then be expressed in a suitable host to produce a polypeptide comprising the peptide or variant of the invention. Thus, the DNA encoding the peptide for use of the invention may be used in accordance with known techniques, appropriately modified in view of the teachings contained herein, to construct an expression vector, which is then used to transform an appropriate host cell for the expression and production of the polypeptide of the invention. Such techniques include those disclosed, for example, in US 4,440,859 , 4,530,901 , 4,582,800 , 4,677,063 , 4,678,751 , 4,704,362 , 4,710,463 , 4,757,006 , 4,766,075 , and 4,810,648 .
  • The DNA (or in the case of retroviral vectors, RNA) encoding the peptide for use of the invention may be joined to a wide variety of other DNA sequences for introduction into an appropriate host. The companion DNA will depend upon the nature of the host, the manner of the introduction of the DNA into the host, and whether episomal maintenance or integration is desired.
  • Generally, the DNA is inserted into an expression vector, such as a plasmid, in proper orientation and correct reading frame for expression. If necessary, the DNA may be linked to the appropriate transcriptional and translational regulatory control nucleotide sequences recognized by the desired host, although such controls are generally available in the expression vector. The vector is then introduced into the host through standard techniques. Generally, not all of the hosts will be transformed by the vector. Therefore, it will be necessary to select for transformed host cells. One selection technique involves incorporating into the expression vector a DNA sequence, with any necessary control elements, that codes for a selectable trait in the transformed cell, such as antibiotic resistance.
  • Alternatively, the gene for such selectable trait can be on another vector, which is used to co-transform the desired host cell.
  • Host cells that have been transformed by the recombinant DNA of the invention are then cultured for a sufficient time and under appropriate conditions known to those skilled in the art in view of the teachings disclosed herein to permit the expression of the polypeptide, which can then be recovered.
  • Many expression systems are known, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae), filamentous fungi (for example Aspergillus spec.), plant cells, animal cells and insect cells. Preferably, the system can be mammalian cells such as CHO cells available from the ATCC Cell Biology Collection.
  • A typical mammalian cell vector plasmid for constitutive expression comprises the CMV or SV40 promoter with a suitable poly A tail and a resistance marker, such as neomycin. One example is pSVL available from Pharmacia, Piscataway, NJ, USA. An example of an inducible mammalian expression vector is pMSG, also available from Pharmacia. Useful yeast plasmid vectors are pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA. Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, TRP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps). CMV promoter-based vectors (for example from Sigma-Aldrich) provide transient or stable expression, cytoplasmic expression or secretion, and N-terminal or C-terminal tagging in various combinations of FLAG, 3xFLAG, c-myc or MAT. These fusion proteins allow for detection, purification and analysis of recombinant protein. Dual-tagged fusions provide flexibility in detection.
  • The strong human cytomegalovirus (CMV) promoter regulatory region drives constitutive protein expression levels as high as 1 mg/L in COS cells. For less potent cell lines, protein levels are typically ∼0.1 mg/L. The presence of the SV40 replication origin will result in high levels of DNA replication in SV40 replication permissive COS cells. CMV vectors, for example, can contain the pMB1 (derivative of pBR322) origin for replication in bacterial cells, the b-lactamase gene for ampicillin resistance selection in bacteria, hGH polyA, and the f1 origin. Vectors containing the pre-pro-trypsin leader (PPT) sequence can direct the secretion of FLAG fusion proteins into the culture medium for purification using ANTI-FLAG antibodies, resins, and plates. Other vectors and expression systems are well known in the art for use with a variety of host cells.
  • In another embodiment two or more peptides as disclosed are encoded and thus expressed in a successive order (similar to "beads on a string" constructs). In doing so, the peptides or peptide variants may be linked or fused together by stretches of linker amino acids, such as for example LLLLLL, or may be linked without any additional peptide(s) between them. These constructs can also be used for cancer therapy, and may induce immune responses both involving MHC I and MHC II.
  • The present invention also relates to a host cell transformed with a polynucleotide vector construct of the present invention. The host cell can be either prokaryotic or eukaryotic. Bacterial cells may be preferred prokaryotic host cells in some circumstances and typically are a strain of E. coli such as, for example, the E. coli strains DH5 available from Bethesda Research Laboratories Inc., Bethesda, MD, USA, and RR1 available from the American Type Culture Collection (ATCC) of Rockville, MD, USA (No ATCC 31343). Preferred eukaryotic host cells include yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human fibroblastic and colon cell lines. Yeast host cells include YPH499, YPH500 and YPH501, which are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA. Preferred mammalian host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells NIH/3T3 available from the ATCC as CRL 1658, monkey kidney-derived COS-1 cells available from the ATCC as CRL 1650 and 293 cells which are human embryonic kidney cells. Preferred insect cells are Sf9 cells which can be transfected with baculovirus expression vectors. An overview regarding the choice of suitable host cells for expression can be found in, for example, the textbook of Paulina Balbás and Argelia Lorence "Methods in Molecular Biology Recombinant Gene Expression, Reviews and Protocols," Part One,
  • Second Edition, ISBN 978-1-58829-262-9, and other literature known to the person of skill.
  • Transformation of appropriate cell hosts with a DNA construct of the present invention is accomplished by well-known methods that typically depend on the type of vector used. With regard to transformation of prokaryotic host cells, see, for example, Cohen et al. (Cohen et al., 1972) and (Green and Sambrook, 2012) . Transformation of yeast cells is described in Sherman et al. (Sherman et al., 1986). The method of Beggs (Beggs, 1978) is also useful. With regard to vertebrate cells, reagents useful in transfecting such cells, for example calcium phosphate and DEAE-dextran or liposome formulations, are available from Stratagene Cloning Systems, or Life Technologies Inc., Gaithersburg, MD 20877, USA. Electroporation is also useful for transforming and/or transfecting cells and is well known in the art for transforming yeast cell, bacterial cells, insect cells and vertebrate cells.
  • Successfully transformed cells, i.e. cells that contain a DNA construct of the present invention, can be identified by well-known techniques such as PCR. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
  • It will be appreciated that certain host cells of the invention are useful in the preparation of the peptide for use of the invention, for example bacterial, yeast and insect cells. However, other host cells may be useful in certain therapeutic methods. For example, antigen-presenting cells, such as dendritic cells, may usefully be used to express the peptide for use of the invention such that they may be loaded into appropriate MHC molecules. Thus, the current invention provides a host cell comprising a nucleic acid or an expression vector according to the invention.
  • In a preferred embodiment the host cell is an antigen presenting cell, in particular a dendritic cell or antigen presenting cell. APCs loaded with a recombinant fusion protein containing prostatic acid phosphatase (PAP) were approved by the U.S. Food and Drug Administration (FDA) on April 29, 2010, to treat asymptomatic or minimally symptomatic metastatic HRPC (Sipuleucel-T) (Rini et al., 2006; Small et al., 2006).
  • A further aspect of the invention provides a method of producing a peptide for use, the method comprising culturing a host cell and isolating the peptide from the host cell or its culture medium.
  • The peptide, the nucleic acid or the expression vector of the invention are used in medicine. For example, the peptide may be prepared for intravenous (i.v.) injection, sub-cutaneous (s.c.) injection, intradermal (i.d.) injection, intraperitoneal (i.p.) injection, intramuscular (i.m.) injection. Preferred methods of peptide injection include s.c., i.d., i.p., i.m., and i.v. Preferred methods of DNA injection include i.d., i.m., s.c., i.p. and i.v. Doses of e.g. between 50 µg and 1.5 mg, preferably 125 µg to 500 µg, of peptide or DNA may be given and will depend on the respective peptide or DNA. Dosages of this range were successfully used in previous trials (Walter et al., 2012).
  • The polynucleotide used for active vaccination may be substantially pure, or contained in a suitable vector or delivery system. The nucleic acid may be DNA, cDNA, PNA, RNA or a combination thereof. Methods for designing and introducing such a nucleic acid are well known in the art. An overview is provided by e.g. Teufel et al. (Teufel et al., 2005). Polynucleotide vaccines are easy to prepare, but the mode of action of these vectors in inducing an immune response is not fully understood. Suitable vectors and delivery systems include viral DNA and/or RNA, such as systems based on adenovirus, vaccinia virus, retroviruses, herpes virus, adeno-associated virus or hybrids containing elements of more than one virus. Non-viral delivery systems include cationic lipids and cationic polymers and are well known in the art of DNA delivery. Physical delivery, such as via a "gene-gun" may also be used. The peptide or peptides encoded by the nucleic acid may be a fusion protein, for example with an epitope that stimulates T cells for the respective opposite CDR as noted above.
  • The medicament for use of the invention may also include one or more adjuvants. Adjuvants are substances that non-specifically enhance or potentiate the immune response (e.g., immune responses mediated by CD8-positive T cells and helper-T (TH) cells to an antigen, and would thus be considered useful in the medicament of the present invention. Suitable adjuvants include, but are not limited to, 1018 ISS, aluminum salts, AMPLIVAX®, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, flagellin or TLR5 ligands derived from flagellin, FLT3 ligand, GM-CSF, IC30, IC31, Imiquimod (ALDARA®), resiquimod, ImuFact IMP321, Interleukins as IL-2, IL-13, IL-21, Interferon-alpha or -beta, or pegylated derivatives thereof, IS Patch, ISS, ISCOMATRIX, ISCOMs, JuvImmune®, LipoVac, MALP2, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, water-in-oil and oil-in-water emulsions, OK-432, OM-174, OM-197-MP-EC, ONTAK, OspA, PepTel® vector system, poly(lactid co-glycolid) [PLG]-based and dextran microparticles, talactoferrin SRL172, Virosomes and other Virus-like particles, YF-17D, VEGF trap, R848, beta-glucan, Pam3Cys, Aquila's QS21 stimulon, which is derived from saponin, mycobacterial extracts and synthetic bacterial cell wall mimics, and other proprietary adjuvants such as Ribi's Detox, Quil, or Superfos. Adjuvants such as Freund's or GM-CSF are preferred. Several immunological adjuvants (e.g., MF59) specific for dendritic cells and their preparation have been described previously (Allison and Krummel, 1995). Also cytokines may be used. Several cytokines have been directly linked to influencing dendritic cell migration to lymphoid tissues (e.g., TNF-), accelerating the maturation of dendritic cells into efficient antigen-presenting cells for T-lymphocytes (e.g., GM-CSF, IL-1 and IL-4) ( U.S. Pat. No. 5,849,589 ) and acting as immunoadjuvants (e.g., IL-12, IL-15, IL-23, IL-7, IFN-alpha. IFN-beta) (Gabrilovich et al., 1996).
  • CpG immunostimulatory oligonucleotides have also been reported to enhance the effects of adjuvants in a vaccine setting. Without being bound by theory, CpG oligonucleotides act by activating the innate (non-adaptive) immune system via Toll-like receptors (TLR), mainly TLR9. CpG triggered TLR9 activation enhances antigen-specific humoral and cellular responses to a wide variety of antigens, including peptide or protein antigens, live or killed viruses, dendritic cell vaccines, autologous cellular vaccines and polysaccharide conjugates in both prophylactic and therapeutic vaccines. More importantly it enhances dendritic cell maturation and differentiation, resulting in enhanced activation of TH1 cells and strong cytotoxic T-lymphocyte (CTL) generation, even in the absence of CD4 T cell help. The TH1 bias induced by TLR9 stimulation is maintained even in the presence of vaccine adjuvants such as alum or incomplete Freund's adjuvant (IFA) that normally promote a TH2 bias. CpG oligonucleotides show even greater adjuvant activity when formulated or co-administered with other adjuvants or in formulations such as microparticles, nanoparticles, lipid emulsions or similar formulations, which are especially necessary for inducing a strong response when the antigen is relatively weak. They also accelerate the immune response and enable the antigen doses to be reduced by approximately two orders of magnitude, with comparable antibody responses to the full-dose vaccine without CpG in some experiments (Krieg, 2006). US 6,406,705 B1 describes the combined use of CpG oligonucleotides, non-nucleic acid adjuvants and an antigen to induce an antigen-specific immune response. A CpG TLR9 antagonist is dSLIM (double Stem Loop Immunomodulator) by Mologen (Berlin, Germany) which is a preferred component of the pharmaceutical composition of the present invention. Other TLR binding molecules such as RNA binding TLR 7, TLR 8 and/or TLR 9 may also be used.
  • Other examples for useful adjuvants include, but are not limited to chemically modified CpGs (e.g. CpR, Idera), dsRNA analogues such as Poly(I:C) and derivates thereof (e.g. AmpliGen®, Hiltonol®, poly-(ICLC), poly(IC-R), poly(I:C12U), non-CpG bacterial DNA or RNA as well as immunoactive small molecules and antibodies such as cyclophosphamide, sunitinib, Bevacizumab®, celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafenib, temozolomide, temsirolimus, XL-999, CP-547632, pazopanib, VEGF Trap, ZD2171, AZD2171, anti-CTLA4, other antibodies targeting key structures of the immune system (e.g. anti-CD40, anti-TGFbeta, anti-TNFalpha receptor) and SC58175, which may act therapeutically and/or as an adjuvant. The amounts and concentrations of adjuvants and additives useful in the context of the present invention can readily be determined by the skilled artisan without undue experimentation.
  • Preferred adjuvants are anti-CD40, imiquimod, resiquimod, GM-CSF, cyclophosphamide, sunitinib, bevacizumab, interferon-alpha, CpG oligonucleotides and derivates, poly-(I:C) and derivates, RNA, sildenafil, and particulate formulations with PLG or virosomes.
  • In a preferred embodiment, the pharmaceutical composition for use according to the invention the adjuvant is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod, resiquimod, and interferon-alpha.
  • In a preferred embodiment, the pharmaceutical composition for use according to the invention the adjuvant is selected from the group consisting of colony-stimulating factors, such as Granulocyte Macrophage Colony Stimulating Factor (GM-CSF, sargramostim), cyclophosphamide, imiquimod and resiquimod. In a preferred embodiment of the pharmaceutical composition for use according to the invention, the adjuvant is cyclophosphamide, imiquimod or resiquimod. Even more preferred adjuvants are Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, poly-ICLC (Hiltonol®) and anti-CD40 mAB, or combinations thereof.
  • This composition is used for parenteral administration, such as subcutaneous, intradermal, intramuscular or oral administration. For this, the peptides and optionally other molecules are dissolved or suspended in a pharmaceutically acceptable, preferably aqueous carrier. In addition, the composition can contain excipients, such as buffers, binding agents, blasting agents, diluents, flavors, lubricants, etc. The peptides can also be administered together with immune stimulating substances, such as cytokines. An extensive listing of excipients that can be used in such a composition, can be, for example, taken from A. Kibbe, Handbook of Pharmaceutical Excipients (Kibbe, 2000). The composition can be used for a prevention, prophylaxis and/or therapy of adenomatous or cancerous diseases. Exemplary formulations can be found in, for example, EP2112253 .
  • It is important to realize that the immune response triggered by the vaccine for use according to the invention attacks the cancer in different cell-stages and different stages of development. Furthermore, different cancer associated signaling pathways are attacked. This is an advantage over vaccines that address only one or few targets, which may cause the tumor to easily adapt to the attack (tumor escape). Furthermore, not all individual tumors express the same pattern of antigens. Therefore, a combination of several tumor-associated peptides ensures that every single tumor bears at least some of the targets. The composition is designed in such a way that each tumor is expected to express several of the antigens and cover several independent pathways necessary for tumor growth and maintenance. Thus, the vaccine can easily be used "off-the-shelf" for a larger patient population. This means that a pre-selection of patients to be treated with the vaccine can be restricted to HLA typing, does not require any additional biomarker assessments for antigen expression, but it is still ensured that several targets are simultaneously attacked by the induced immune response, which is important for efficacy (Banchereau et al., 2001; Walter et al., 2012).
  • The peptide for use of the present invention can be used to generate and develop specific antibodies against MHC/peptide complexes. These can be used for therapy, targeting toxins or radioactive substances to the diseased tissue. Another use of these antibodies can be targeting radionuclides to the diseased tissue for imaging purposes such as PET. This use can help to detect small metastases or to determine the size and precise localization of diseased tissues.
  • Therefore, it is a further aspect of the invention to provide a method for producing a recombinant antibody specifically binding to a human major histocompatibility complex (MHC) class I being complexed with a HLA-restricted antigen, the method comprising: immunizing a genetically engineered non-human mammal comprising cells expressing said human major histocompatibility complex (MHC) class I with a soluble form of a MHC class I molecule being complexed with said HLA-restricted antigen; isolating mRNA molecules from antibody producing cells of said non-human mammal; producing a phage display library displaying protein molecules encoded by said mRNA molecules; and isolating at least one phage from said phage display library, said at least one phage displaying said antibody specifically binding to said human major histocompatibility complex (MHC) class I being complexed with said HLA-restricted antigen.
  • It is a further aspect of the invention to provide an antibody that specifically binds to a human major histocompatibility complex (MHC) class I being complexed with a HLA-restricted antigen, wherein the antibody preferably is a polyclonal antibody, monoclonal antibody, bi-specific antibody and/or a chimeric antibody.
  • Respective methods for producing such antibodies and single chain class I major histocompatibility complexes, as well as other tools for the production of these antibodies are disclosed in WO 03/068201 , WO 2004/084798 , WO 01/72768 , WO 03/070752 , and in publications (Cohen et al., 2003a; Cohen et al., 2003b; Denkberg et al., 2003).
  • Preferably, the antibody is binding with a binding affinity of below 20 nanomolar, preferably of below 10 nanomolar, to the complex, which is also regarded as "specific" in the context of the present invention.
  • The present invention relates to peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  • The present invention further relates to the peptide for use according to the invention that has the ability to bind to a molecule of the human major histocompatibility complex (MHC) class-I.
  • The present invention further relates to the peptide for use according to the invention, wherein the peptide includes non-peptide bonds.
  • The present invention further relates to the peptides according to the invention, wherein the peptide is part of a fusion protein, comprising the N-terminal 80 amino acids of the HLA-DR antigen-associated invariant chain (li).
  • Another embodiment of the present invention relates to a non-naturally occurring peptide for use wherein said peptide consists of the amino acid sequence according to SEQ ID No: 9 and has been synthetically produced (e.g. synthesized) as a pharmaceutically acceptable salt. Methods to synthetically produce peptides are well known in the art. The salts of the peptide for use according to the present invention differ substantially from the peptide in its state in vivo, as the peptide as generated in vivo is no salt. The non-natural salt form of the peptide mediates the solubility of the peptide, in particular in the context of pharmaceutical compositions comprising the peptides for use, e.g. the peptide vaccines for use as disclosed herein. A sufficient and at least substantial solubility of the peptide(s) is required in order to efficiently provide the peptides to the subject to be treated. Preferably, the salts are pharmaceutically acceptable salts of the peptide for use. These salts according to the invention include alkaline and earth alkaline salts such as salts of the Hofmeister series comprising as anions PO4 3-, SO4 2-, CH3COO-, Cl-, Br-, NO3 -, ClO4 -, I-, SCN- and as cations NH4 +, Rb+, K+, Na+, Cs+, Li+, Zn2+, Mg2+, Ca2+, Mn2+, Cu2+ and Ba2+. Particularly salts are selected from (NH4)3PO4, (NH4)2HPO4, (NH4)H2PO4, (NH4)2SO4, NH4CH3COO, NH4Cl, NH4Br, NH4NO3, NH4ClO4, NH4I, NH4SCN, Rb3PO4, Rb2HPO4, RbH2PO4, Rb2SO4, Rb4CH3COO, Rb4Cl, Rb4Br, Rb4NO3, Rb4ClO4, Rb4I, Rb4SCN, K3PO4, K2HPO4, KH2PO4, K2SO4, KCH3COO, KCl, KBr, KNO3, KClO4, KI, KSCN, Na3PO4, Na2HPO4, NaH2PO4, Na2SO4, NaCH3COO, NaCl, NaBr, NaNO3, NaClO4, Nal, NaSCN, ZnCl2 Cs3PO4, Cs2HPO4, CsH2PO4, Cs2SO4, CsCH3COO, CsCl, CsBr, CsNO3, CsClO4, Csl, CsSCN, Li3PO4, Li2HPO4, LiH2PO4, Li2SO4, LiCH3COO, LiCl, LiBr, LiNO3, LiClO4, Lil, LiSCN, Cu2SO4, Mg3(PO4)2, Mg2HPO4, Mg(H2PO4)2, Mg2SO4, Mg(CH3COO)2, MgCl2, MgBr2, Mg(NO3)2, Mg(ClO4)2, MgI2, Mg(SCN)2, MnCl2, Ca3(PO4),, Ca2HPO4, Ca(H2PO4)2, CaSO4, Ca(CH3COO)2, CaCl2, CaBr2, Ca(NO3)2, Ca(ClO4)2, CaI2, Ca(SCN)2, Ba3(PO4)2, Ba2HPO4, Ba(H2PO4)2, BaSO4, Ba(CH3COO)2, BaCl2, BaBr2, Ba(NO3)2, Ba(ClO4)2, BaI2, and Ba(SCN)2. Particularly preferred are NH acetate, MgCl2, KH2PO4, Na2SO4, KCl, NaCl, and CaCl2, such as, for example, the chloride or acetate (trifluoroacetate) salts.
  • Generally, peptides (at least those containing peptide linkages between amino acid residues) may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis as disclosed by Lukas et al. (Lukas et al., 1981) and by references as cited therein. Temporary N-amino group protection is afforded by the 9-fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide. Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine). Where glutamine or asparagine are C-terminal residues, use is made of the 4,4'-dimethoxybenzhydryl group for protection of the side chain amido functionalities. The solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalizing agent). The peptide-to-resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives with the exception of asparagine and glutamine, which are added using a reversed N, N-dicyclohexylcarbodiimide/1hydroxybenzotriazole mediated coupling procedure. All coupling and deprotection reactions are monitored using ninhydrine, trinitrobenzene sulphonic acid or isotin test procedures. Upon completion of synthesis, peptides are cleaved from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50 % scavenger mix. Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible (see, for example, (Bruckdorfer et al., 2004), and the references as cited therein).
  • Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide. Any scavengers present are removed by a simple extraction procedure which on lyophilization of the aqueous phase affords the crude peptide free of scavengers. Reagents for peptide synthesis are generally available from e.g. Calbiochem-Novabiochem (Nottingham, UK).
  • Purification may be performed by any one, or a combination of, techniques such as recrystallization, size exclusion chromatography, ion-exchange chromatography, hydrophobic interaction chromatography and (usually) reverse-phase high performance liquid chromatography using e.g. acetonitril/water gradient separation.
  • The present invention further relates to a nucleic acid, encoding the peptide for use according to the invention.
  • The present invention further relates to the nucleic acid according to the invention that is DNA, cDNA, PNA, RNA or combinations thereof.
  • The present invention further relates to an expression vector expressing a nucleic acid according to the present invention.
  • The present invention further relates to a peptide according to the present invention, a nucleic acid according to the present invention or an expression vector according to the present invention for use in medicine, in particular in the treatment of esophageal cancer.
  • The present invention further relates to a host cell comprising a nucleic acid according to the invention or an expression vector according to the invention.
  • The present invention further relates to the host cell according to the present invention that is an antigen presenting cell, and preferably a dendritic cell.
  • The present invention further relates to a method of producing a peptide for use according to the present invention, said method comprising culturing the host cell according to the present invention, and isolating the peptide from said host cell or its culture medium.
  • The present invention further relates to the method according to the present invention, wherein the antigen is loaded onto class I MHC molecules expressed on the surface of a suitable antigen-presenting cell by contacting a sufficient amount of the antigen with an antigen-presenting cell.
  • The present invention further relates to the method according to the invention, wherein the antigen-presenting cell comprises an expression vector expressing said peptide containing SEQ ID NO: 9.
  • The present invention further relates to activated T cells, produced by the method according to the present invention, wherein said T cells selectively recognizes a cell which aberrantly expresses a polypeptide comprising an amino acid sequence for use according to the present invention.
  • Disclosed is a method of killing target cells in a patient which target cells aberrantly express a polypeptide comprising any amino acid sequence according to the present invention, the method comprising administering to the patient an effective number of T cells as according to the present invention.
  • Disclosed is the use of any peptide described, a nucleic acid according to the present invention, an expression vector according to the present invention, a cell according to the present invention, or an activated cytotoxic T lymphocyte according to the present invention as a medicament or in the manufacture of a medicament. The present invention further relates to a use according to the present invention, wherein the medicament is active against cancer.
  • The present invention further relates to a use according to the invention, wherein the medicament is a vaccine. The present invention further relates to a use according to the invention, wherein the medicament is active against cancer.
  • The present invention further relates to a use according to the invention, wherein said cancer cells are esophageal cancer cells or other solid or hematological tumor cells such as lung cancer, urinary bladder cancer, ovarian cancer, melanoma, uterine cancer, hepatocellular cancer, renal cell cancer, brain cancer, colorectal cancer, breast cancer, gastric cancer, pancreatic cancer, gallbladder cancer, bile duct cancer, prostate cancer and leukemia.
  • The term "antibody" or "antibodies" is used herein in a broad sense and includes both polyclonal and monoclonal antibodies. In addition to intact or "full" immunoglobulin molecules, also included in the term "antibodies" are fragments (e.g. CDRs, Fv, Fab and Fc fragments) or polymers of those immunoglobulin molecules and humanized versions of immunoglobulin molecules, as long as they exhibit any of the desired properties (e.g., specific binding of a esophageal cancer marker (poly)peptide, delivery of a toxin to a esophageal cancer cell expressing a cancer marker gene at an increased level, and/or inhibiting the activity of a esophageal cancer marker polypeptide) according to the invention.
  • Whenever possible, the antibodies of the invention may be purchased from commercial sources. The antibodies of the invention may also be generated using well-known methods. The skilled artisan will understand that either full length esophageal cancer marker polypeptides or fragments thereof may be used to generate the antibodies of the invention. A polypeptide to be used for generating an antibody of the invention may be partially or fully purified from a natural source, or may be produced using recombinant DNA techniques.
  • For example, a cDNA encoding a peptide for use according to the present invention, such as a peptide according to SEQ ID NO: 9, can be expressed in prokaryotic cells (e.g., bacteria) or eukaryotic cells (e.g., yeast, insect, or mammalian cells), after which the recombinant protein can be purified and used to generate a monoclonal or polyclonal antibody preparation that specifically bind the esophageal cancer marker polypeptide used to generate the antibody according to the invention.
  • One of skill in the art will realize that the generation of two or more different sets of monoclonal or polyclonal antibodies maximizes the likelihood of obtaining an antibody with the specificity and affinity required for its intended use (e.g., ELISA, immunohistochemistry, in vivo imaging, immunotoxin therapy). The antibodies are tested for their desired activity by known methods, in accordance with the purpose for which the antibodies are to be used (e.g., ELISA, immunohistochemistry, immunotherapy, etc.; for further guidance on the generation and testing of antibodies, see, e.g., Greenfield, 2014 (Greenfield, 2014)). For example, the antibodies may be tested in ELISA assays or, Western blots, immunohistochemical staining of formalin-fixed cancers or frozen tissue sections. After their initial in vitro characterization, antibodies intended for therapeutic or in vivo diagnostic use are tested according to known clinical testing methods.
  • The term "monoclonal antibody" as used herein refers to an antibody obtained from a substantially homogeneous population of antibodies, i.e.; the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. The monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired antagonistic activity ( US 4,816,567 ).
  • Monoclonal antibodies of the invention may be prepared using hybridoma methods. In a hybridoma method, a mouse or other appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes may be immunized in vitro.
  • The monoclonal antibodies may also be made by recombinant DNA methods, such as those described in US 4,816,567 . DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments thereof, particularly Fab fragments, can be accomplished using routine techniques known in the art. For instance, digestion can be performed using papain. Examples of papain digestion are described in WO 94/29348 and US 4,342,566 . Papain digestion of antibodies typically produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual Fc fragment. Pepsin treatment yields a F(ab')2 fragment and a pFc' fragment.
  • The antibody fragments, whether attached to other sequences or not, can also include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the non-modified antibody or antibody fragment. These modifications can provide for some additional property, such as to remove/add amino acids capable of disulfide bonding, to increase its bio-longevity, to alter its secretory characteristics, etc. In any case, the antibody fragment must possess a bioactive property, such as binding activity, regulation of binding at the binding domain, etc. Functional or active regions of the antibody may be identified by mutagenesis of a specific region of the protein, followed by expression and testing of the expressed polypeptide. Such methods are readily apparent to a skilled practitioner in the art and can include site-specific mutagenesis of the nucleic acid encoding the antibody fragment.
  • The antibodies of the invention may further comprise humanized antibodies or human antibodies. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab' or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies ( US 4,816,567 ), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • Transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production can be employed. For example, it has been described that the homozygous deletion of the antibody heavy chain joining region gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. Human antibodies can also be produced in phage display libraries.
  • Antibodies of the invention are preferably administered to a subject in a pharmaceutically acceptable carrier. Typically, an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic. Examples of the pharmaceutically-acceptable carrier include saline, Ringer's solution and dextrose solution. The pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5. Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of antibody being administered.
  • The antibodies can be administered to the subject, patient, or cell by injection (e.g., intravenous, intraperitoneal, subcutaneous, intramuscular), or by other methods such as infusion that ensure its delivery to the bloodstream in an effective form. The antibodies may also be administered by intratumoral or peritumoral routes, to exert local as well as systemic therapeutic effects. Local or intravenous injection is preferred.
  • Effective dosages and schedules for administering the antibodies may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of antibodies that must be administered will vary depending on, for example, the subject that will receive the antibody, the route of administration, the particular type of antibody used and other drugs being administered. A typical daily dosage of the antibody used alone might range from about 1 (µg/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above. Following administration of an antibody, preferably for treating esophageal cancer, the efficacy of the therapeutic antibody can be assessed in various ways well known to the skilled practitioner. For instance, the size, number, and/or distribution of cancer in a subject receiving treatment may be monitored using standard tumor imaging techniques. A therapeutically-administered antibody that arrests tumor growth, results in tumor shrinkage, and/or prevents the development of new tumors, compared to the disease course that would occur in the absence of antibody administration, is an efficacious antibody for treatment of cancer.
  • It is a further aspect of the invention to provide a method for producing a soluble T-cell receptor (sTCR) recognizing a specific peptide-MHC complex. Such soluble T-cell receptors can be generated from specific T-cell clones, and their affinity can be increased by mutagenesis targeting the complementarity-determining regions. For the purpose of T-cell receptor selection, phage display can be used ( US 2010/0113300, (Liddy et al., 2012 )). For the purpose of stabilization of T-cell receptors during phage display and in case of practical use as drug, alpha and beta chain can be linked e.g. by non-native disulfide bonds, other covalent bonds (single-chain T-cell receptor), or by dimerization domains (Boulter et al., 2003; Card et al., 2004; Willcox et al., 1999). The T-cell receptor can be linked to toxins, drugs, cytokines (see, for example, US 2013/0115191 ), domains recruiting effector cells such as an anti-CD3 domain, etc., in order to execute particular functions on target cells. Moreover, it could be expressed in T cells used for adoptive transfer. Further information can be found in WO 2004/033685A1 and WO 2004/074322A1 . A combination of sTCRs is described in WO 2012/056407A1 . Further methods for the production are disclosed in WO 2013/057586A1 .
  • In addition, the peptides and/or the TCRs or antibodies or other binding molecules of the present invention can be used to verify a pathologist's diagnosis of a cancer based on a biopsied sample.
  • The antibodies or TCRs may also be used for in vivo diagnostic assays. Generally, the antibody is labeled with a radionucleotide (such as 111In, 99Tc, 14C, 131I, 3H, 32P or 35S) so that the tumor can be localized using immunoscintiography. In one embodiment, antibodies or fragments thereof bind to the extracellular domains of two or more targets of a protein selected from the group consisting of the above-mentioned proteins, and the affinity value (Kd) is less than 1 x 10µM.
  • Antibodies for diagnostic use may be labeled with probes suitable for detection by various imaging methods. Methods for detection of probes include, but are not limited to, fluorescence, light, confocal and electron microscopy; magnetic resonance imaging and spectroscopy; fluoroscopy, computed tomography and positron emission tomography. Suitable probes include, but are not limited to, fluorescein, rhodamine, eosin and other fluorophores, radioisotopes, gold, gadolinium and other lanthanides, paramagnetic iron, fluorine-18 and other positron-emitting radionuclides. Additionally, probes may be bi- or multi-functional and be detectable by more than one of the methods listed. These antibodies may be directly or indirectly labeled with said probes. Attachment of probes to the antibodies includes covalent attachment of the probe, incorporation of the probe into the antibody, and the covalent attachment of a chelating compound for binding of probe, amongst others well recognized in the art. For immunohistochemistry, the disease tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin. The fixed or embedded section contains the sample are contacted with a labeled primary antibody and secondary antibody, wherein the antibody is used to detect the expression of the proteins in situ.
  • Another aspect of the present invention includes an in vitro method for producing activated T cells, the method comprising contacting in vitro T cells with antigen loaded human MHC molecules expressed on the surface of a suitable antigen-presenting cell for a period of time sufficient to activate the T cell in an antigen specific manner, wherein the antigen is a peptide according to the invention. Preferably a sufficient amount of the antigen is used with an antigen-presenting cell.
  • Preferably the mammalian cell lacks or has a reduced level or function of the TAP peptide transporter. Suitable cells that lack the TAP peptide transporter include T2, RMA-S and Drosophila cells. TAP is the transporter associated with antigen processing.
  • The human peptide loading deficient cell line T2 is available from the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852, USA under Catalogue No CRL 1992; the Drosophila cell line Schneider line 2 is available from the ATCC under Catalogue No CRL 19863; the mouse RMA-S cell line is described in Ljunggren et al. (Ljunggren and Karre, 1985).
  • Preferably, before transfection the host cell expresses substantially no MHC class I molecules. It is also preferred that the stimulator cell expresses a molecule important for providing a co-stimulatory signal for T-cells such as any of B7.1, B7.2, ICAM-1 and LFA 3. The nucleic acid sequences of numerous MHC class I molecules and of the co-stimulator molecules are publicly available from the GenBank and EMBL databases.
  • In case of a MHC class I epitope being used as an antigen, the T cells are CD8-positive T cells.
  • If an antigen-presenting cell is transfected to express such an epitope, preferably the cell comprises an expression vector expressing a peptide containing SEQ ID NO: 9.
  • A number of other methods may be used for generating T cells in vitro. For example, autologous tumor-infiltrating lymphocytes can be used in the generation of CTL. Plebanski et al. (Plebanski et al., 1995) made use of autologous peripheral blood lymphocytes (PLBs) in the preparation of T cells. Furthermore, the production of autologous T cells by pulsing dendritic cells with peptide or polypeptide, or via infection with recombinant virus is possible. Also, B cells can be used in the production of autologous T cells. In addition, macrophages pulsed with peptide or polypeptide, or infected with recombinant virus, may be used in the preparation of autologous T cells. S. Walter et al. (Walter et al., 2003) describe the in vitro priming of T cells by using artificial antigen presenting cells (aAPCs), which is also a suitable way for generating T cells against the peptide of choice. In the present invention, aAPCs were generated by the coupling of preformed MHC:peptide complexes to the surface of polystyrene particles (microbeads) by biotin:streptavidin biochemistry. This system permits the exact control of the MHC density on aAPCs, which allows to selectively elicit high- or low-avidity antigen-specific T cell responses with high efficiency from blood samples. Apart from MHC:peptide complexes, aAPCs should carry other proteins with co-stimulatory activity like anti-CD28 antibodies coupled to their surface. Furthermore, such aAPC-based systems often require the addition of appropriate soluble factors, e. g. cytokines, like interleukin-12.
  • Allogeneic cells may also be used in the preparation of T cells and a method is described in detail in WO 97/26328 . For example, in addition to Drosophila cells and T2 cells, other cells may be used to present antigens such as CHO cells, baculovirus-infected insect cells, bacteria, yeast, vaccinia-infected target cells. In addition, plant viruses may be used (see, for example, Porta et al. (Porta et al., 1994) which describes the development of cowpea mosaic virus as a high-yielding system for the presentation of foreign peptides.
  • The activated T cells that are directed against the peptide for use of the invention are useful in therapy. Thus, a further aspect of the invention provides activated T cells obtainable by the foregoing methods of the invention.
  • Activated T cells, which are produced by the above method, will selectively recognize a cell that aberrantly expresses a polypeptide that comprises an amino acid sequence of SEQ ID NO: 9.
  • Preferably, the T cell recognizes the cell by interacting through its TCR with the HLA/peptide-complex (for example, binding). The T cells are useful in a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention wherein the patient is administered an effective number of the activated T cells. The T cells that are administered to the patient may be derived from the patient and activated as described above (i.e. they are autologous T cells). Alternatively, the T cells are not from the patient but are from another individual. Of course, it is preferred if the individual is a healthy individual. By "healthy individual" the inventors mean that the individual is generally in good health, preferably has a competent immune system and, more preferably, is not suffering from any disease that can be readily tested for, and detected.
  • In vivo, the target cells for the CD8-positive T cells according to the present invention can be cells of the tumor (which sometimes express MHC class II) and/or stromal cells surrounding the tumor (tumor cells) (which sometimes also express MHC class II; (Dengjel et al., 2006)).
  • The T cells of the present invention may be used as active ingredients of a therapeutic composition. Thus, disclosed is a method of killing target cells in a patient whose target cells aberrantly express a polypeptide comprising an amino acid sequence of the invention, the method comprising administering to the patient an effective number of T cells as defined above.
  • By "aberrantly expressed" the inventors also mean that the polypeptide is over-expressed compared to levels of expression in normal (healthy) tissues or that the gene is silent in the tissue from which the tumor is derived but in the tumor it is expressed. By "over-expressed" the inventors mean that the polypeptide is present at a level at least 1.2-fold of that present in normal tissue; preferably at least 2-fold, and more preferably at least 5-fold or 10-fold the level present in normal tissue.
  • T cells may be obtained by methods known in the art, e.g. those described above.
  • Protocols for this so-called adoptive transfer of T cells are well known in the art. Reviews can be found in: Gattioni et al. and Morgan et al. (Gattinoni et al., 2006; Morgan et al., 2006).
  • Another aspect of the present invention includes the use of the peptide for use complexed with MHC to generate a T-cell receptor whose nucleic acid is cloned and is introduced into a host cell, preferably a T cell. This engineered T cell can then be transferred to a patient for therapy of cancer.
  • Any molecule of the invention, i.e. the peptide for use, nucleic acid, antibody, expression vector, cell, activated T cell, T-cell receptor or the nucleic acid encoding it, is useful for the treatment of disorders, characterized by cells escaping an immune response. Therefore, any molecule of the present invention may be used as medicament or in the manufacture of a medicament. The molecule may be used by itself or combined with other molecule(s) of the invention or (a) known molecule(s).
  • Disclosed is a kit comprising:
    1. (a) a container containing a pharmaceutical composition as described above, in solution or in lyophilized form;
    2. (b) optionally a second container containing a diluent or reconstituting solution for the lyophilized formulation; and
    3. (c) optionally, instructions for (i) use of the solution or (ii) reconstitution and/or use of the lyophilized formulation.
  • The kit may further comprise one or more of (iii) a buffer, (iv) a diluent, (v) a filter, (vi) a needle, or (v) a syringe. The container is preferably a bottle, a vial, a syringe or test tube; and it may be a multi-use container. The pharmaceutical composition is preferably lyophilized.
  • Kits as disclosed preferably comprise a lyophilized formulation of the present invention in a suitable container and instructions for its reconstitution and/or use. Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes. The container may be formed from a variety of materials such as glass or plastic. Preferably the kit and/or container contain/s instructions on or associated with the container that indicates directions for reconstitution and/or use. For example, the label may indicate that the lyophilized formulation is to be reconstituted to peptide concentrations as described above. The label may further indicate that the formulation is useful or intended for subcutaneous administration.
  • The container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation. The kit may further comprise a second container comprising a suitable diluent (e.g., sodium bicarbonate solution).
  • Upon mixing of the diluent and the lyophilized formulation, the final peptide concentration in the reconstituted formulation is preferably at least 0.15 mg/mL/peptide (=75 µg) and preferably not more than 3 mg/mL/peptide (=1500 µg). The kit may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Kits as disclosed may have a single container that contains the formulation of the pharmaceutical compositions for use according to the present invention with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have distinct container for each component.
  • Preferably, kits as disclosed include a formulation of the invention packaged for use in combination with the co-administration of a second compound (such as adjuvants (e.g. GM-CSF), a chemotherapeutic agent, a natural product, a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof. The components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient. The components of the kit may be provided in one or more liquid solutions, preferably, an aqueous solution, more preferably, a sterile aqueous solution. The components of the kit may also be provided as solids, which may be converted into liquids by addition of suitable solvents, which are preferably provided in another distinct container.
  • The container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid. Usually, when there is more than one component, the kit will contain a second vial or other container, which allows for separate dosing. The kit may also contain another container for a pharmaceutically acceptable liquid. Preferably, a therapeutic kit will contain an apparatus (e.g., one or more needles, syringes, eye droppers, pipette, etc.), which enables administration of the agents as disclosed that are components of the present kit.
  • The present formulation is one that is suitable for administration of the peptides by any acceptable route such as oral (enteral), nasal, ophthal, subcutaneous, intradermal, intramuscular, intravenous or transdermal. Preferably, the administration is s.c., and most preferably i.d. administration may be by infusion pump.
  • Since the peptide for use of the invention was isolated from esophageal cancer, the medicament of the invention is preferably used to treat esophageal cancer.
  • In one exemplary embodiment, the peptides included in the vaccine are identified by: (a) identifying tumor-associated peptides (TUMAPs) presented by a tumor sample from the individual patient; and (b) selecting at least one peptide identified de novo in (a) and confirming its immunogenicity.
  • Once the peptides for a personalized peptide based vaccine are selected, the vaccine is produced. The vaccine preferably is a liquid formulation consisting of the individual peptides dissolved in between 20-40% DMSO, preferably about 30-35% DMSO, such as about 33% DMSO.
  • Each peptide to be included into a product is dissolved in DMSO. The concentration of the single peptide solutions has to be chosen depending on the number of peptides to be included into the product. The single peptide-DMSO solutions are mixed in equal parts to achieve a solution containing all peptides to be included in the product with a concentration of -2.5 mg/ml per peptide. The mixed solution is then diluted 1:3 with water for injection to achieve a concentration of 0.826 mg/ml per peptide in 33% DMSO. The diluted solution is filtered through a 0.22 µm sterile filter. The final bulk solution is obtained.
  • Final bulk solution is filled into vials and stored at -20°C until use. One vial contains 700 µL solution, containing 0.578 mg of each peptide. Of this, 500 µL (approx. 400 µg per peptide) will be applied for intradermal injection.
  • The present invention will now be described in the following examples which describe preferred embodiments thereof, and with reference to the accompanying figures, nevertheless, without being limited thereto.
  • FIGURES
    • Figure 1A to 1V show the over-presentation of various peptides in normal tissues (white bars) and esophageal cancer (black bars). A) Gene symbol: KRT14/KRT16, Peptide: STYGGGLSV (SEQ ID NO: 1) Tissues from left to right: 1 adipose tissues, 3 adrenal glands, 8 arteries, 5 bone marrows, 7 brains, 5 breasts, 2 cartilages, 1 central nerve , 13 colons, 1 duodenum, 2 gallbladders, 5 hearts, 14 kidneys, 21 livers, 44 lungs, 4 lymph nodes, 4 leukocyte samples, 3 ovaries, 8 pancreas, 5 peripheral nerves, 1 peritoneum, 3 pituitary glands, 4 placentas, 3 pleuras, 3 prostates, 6 recti, 7 salivary glands, 4 skeletal muscles, 6 skins, 2 small intestines, 4 spleens, 5 stomachs, 6 testis, 3 thymi, 3 thyroid glands, 7 tracheas, 2 ureters, 6 urinary bladders, 2 uteri, 2 veins, 6 esophagi, 16 esophageal cancer samples. The peptide has additionally been detected on 4/91 lung cancers. Figure 1B) Gene symbol: GJB5, Peptide: SIFEGLLSGV (SEQ ID NO: 7). Tissues from left to right: 1 adipose tissues, 3 adrenal glands, 8 arteries, 5 bone marrows, 7 brains, 5 breasts, 2 cartilages, 1 central nerve , 13 colons, 1 duodenum, 2 gallbladders, 5 hearts, 14 kidneys, 21 livers, 44 lungs, 4 lymph nodes, 4 leukocyte samples, 3 ovaries, 8 pancreas, 5 peripheral nerves, 1 peritoneum, 3 pituitary glands, 4 placentas, 3 pleuras, 3 prostates, 6 recti, 7 salivary glands, 4 skeletal muscles, 6 skins, 2 small intestines, 4 spleens, 5 stomachs, 6 testis, 3 thymi, 3 thyroid glands, 7 tracheas, 2 ureters, 6 urinary bladders, 2 uteri, 2 veins, 6 esophagi, 16 esophageal cancer samples. The peptide has additionally been detected on 1/43 prostate cancers, 1/3 gallbladder cancers, 1/20 ovarian cancers, 5/91 lung cancers and 1/4 urinary bladder cancers. Figure 2C) Gene symbol: PKP3, Peptide: SLVSEQLEPA (SEQ ID NO: 34). Tissues from left to right: 1 adipose tissues, 3 adrenal glands, 8 arteries, 5 bone marrows, 7 brains, 5 breasts, 2 cartilages, 1 central nerve , 13 colons, 1 duodenum, 2 gallbladders, 5 hearts, 14 kidneys, 21 livers, 44 lungs, 4 lymph nodes, 4 leukocyte samples, 3 ovaries, 8 pancreas, 5 peripheral nerves, 1 peritoneum, 3 pituitary glands, 4 placentas, 3 pleuras, 3 prostates, 6 recti, 7 salivary glands, 4 skeletal muscles, 6 skins, 2 small intestines, 4 spleens, 5 stomachs, 6 testis, 3 thymi, 3 thyroid glands, 7 tracheas, 2 ureters, 6 urinary bladders, 2 uteri, 2 veins, 6 esophagi, 16 esophageal cancer samples. The peptide has additionally been detected on 8/24 colorectal cancers, 1/20 ovarian cancers, 1/46 gastric cancers, 5/91 lung cancers and 2/4 urinary bladder cancers. Figure 3D) Gene symbol: RNPEP, Peptide: YTQPFSHYGQAL (SEQ ID NO: 37). Tissues from left to right: 1 adipose tissues, 3 adrenal glands, 8 arteries, 5 bone marrows, 7 brains, 5 breasts, 2 cartilages, 1 central nerve , 13 colons, 1 duodenum, 2 gallbladders, 5 hearts, 14 kidneys, 21 livers, 44 lungs, 4 lymph nodes, 4 leukocyte samples, 3 ovaries, 8 pancreas, 5 peripheral nerves, 1 peritoneum, 3 pituitary glands, 4 placentas, 3 pleuras, 3 prostates, 6 recti, 7 salivary glands, 4 skeletal muscles, 6 skins, 2 small intestines, 4 spleens, 5 stomachs, 6 testis, 3 thymi, 3 thyroid glands, 7 tracheas, 2 ureters, 6 urinary bladders, 2 uteri, 2 veins, 6 esophagi, 16 esophageal cancer samples. The peptide has additionally been detected on 1/19 pancreatic cancers, 7/46 gastric cancers and 1/91 lung cancers.. Figure 4E) Gene symbol: NUP155, Peptide: ALQEALENA (SEQ ID NO: 80). Samples from left to right: 4 cell lines (1 kidney, 1 pancreatic, 1 prostate, 1 myeloid leukemia), 3 normal tissues (1 lung, 1 prostate, 1 small intestine), 47 cancer tissues (5 brain cancers, 2 breast cancers, 1 colon cancers, 2 esophageal cancers, 1 chronic leukocytic leukemia, 2 liver cancers, 22 lung cancers, 7 ovarian cancers, 4 prostate cancers, 1 rectum cancer). Figure 5F) Gene symbol: KRT5, Peptide: SLYNLGGSKRISI (SEQ ID NO: 2). Tissues from left to right: 20 cancer tissues (9 head-and-neck cancers, 2 esophageal cancers, 1 esophagus and stomach cancer, 7 lung cancers, 1 urinary bladder cancer). Figure 6G) Gene symbol: KRT5, Peptide: TASAITPSV (SEQ ID NO: 3). Tissues from left to right: 17 cancer tissues (2 esophageal cancers, 6 head-and-neck cancers, 7 lung cancers, 2 urinary bladder cancers). Figure 7H) Gene symbol: S100A2, Peptide: SLDENSDQQV (SEQ ID NO: 10). Tissues from left to right: 7 cancer tissues (3 head-and-neck cancers, 2 esophageal cancers, 1 lung cancer, 1 urinary bladder cancer). Figure 8I) Gene symbol: LAMB3, Peptide: ALWLPTDSATV (SEQ ID NO: 11). Tissues from left to right: 12 cancer tissues (2 esophageal cancers, 1 gallbladder cancer, 8 lung cancers, 1 skin cancer). Figure 9J) Gene symbol: IL36RN, Peptide: SLSPVILGV (SEQ ID NO: 13). Tissues from left to right: 26 cancer tissues (8 head-and-neck cancers, 3 esophageal cancers, 10 lung cancers, 3 skin cancers, 1 urinary bladder cancer, 1 uterus cancer). Figure 10K) Gene symbol: ANO1, Peptide: LLANGVYAA (SEQ ID NO: 15). Tissues from left to right: 8 cancer tissues (2 esophageal cancers, 1 gallbladder cancer, 1 liver cancer, 1 lung cancer, 1 stomach cancer, 1 urinary bladder cancer, 1 uterus cancer). Figure 11L) Gene symbol: F7, IGHV4-31, IGHG1, IGHG2, IGHG3, IGHG4, IGHM, Peptide: MISRTPEV (SEQ ID NO: 17). Tissues from left to right: 19 cancer tissues (2 esophageal cancers, 2 kidney cancers, 2 liver cancers, 9 lung cancers, 1 lymph node cancer, 1 testis cancer, 2 urinary bladder cancers. Figure 12M) Gene symbol: QSER1, Peptide: SLNGNQVTV (SEQ ID NO: 30). Tissues from left to right: 1 cell line (1 pancreatic), 14 cancer tissues (1 head-and-neck cancer, 1 bile duct cancer, 1 brain cancer, 1 breast cancer, 1 esophageal cancer, 1 kidney cancer, 1 lung cancer, 2 skin cancers, 3 urinary bladder cancers, 2 uterus cancers). Figure 13N) Gene symbol: HAS3, Peptide: YMLDIFHEV (SEQ ID NO: 32). Tissues from left to right: 1 normal tissue (1 uterus), 15 cancer tissues (1 brain cancer, 2 esophageal cancers, 1 gallbladder cancer, 3 head-and-neck cancers, 4 lung cancers, 4 urinary bladder cancers). Figure 14O) Gene symbol: PKP3, Peptide: SLVSEQLEPA (SEQ ID NO: 34). Tissues from left to right: 1 cell line (1 pancreatic), 1 normal tissue (1 colon), 28 cancer tissues (6 head-and-neck cancers, 1 breast cancer, 1 cecum cancer, 3 colon cancers, 1 colorectal cancer, 3 esophageal cancers, 6 lung cancers, 1 ovarian cancer, 3 rectum cancers, 3 urinary bladder cancers). Figure 15P) Gene symbol: SERPINH1, Peptide: GLAFSLYQA (SEQ ID NO: 40). Tissues from left to right: 3 cell lines (1 kidney, 2 pancreatic), 4 normal tissues (1 adrenal gland, 1 lung, 2 placentas), 41 cancer tissues (3 head-and-neck cancers, 3 breast cancers, 2 colon cancers, 2 esophageal cancers, 1 gallbladder cancer, 1 liver cancer, 15 lung cancers, 1 ovarian cancer, 1 pancreas cancer, 3 rectum cancers, 2 skin cancers, 1 stomach cancer, 4 urinary bladder cancers, 2 uterus cancers). Figure 1Q) Gene symbol: TMEM132A, Peptide: ALVEVTEHV (SEQ ID NO: 56). Tissues from left to right: 7 normal tissues (5 lungs, 1 thyroid gland, 1 trachea), 64 cancer tissues (6 head-and-neck cancers, 12 brain cancers, 4 breast cancers, 3 esophageal cancers, 1 gallbladder cancer, 5 kidney cancers, 21 lung cancers, 1 lymph node cancer, 7 ovarian cancers, 1 pancreas cancer, 1 skin cancer, 2 uterus cancers). Figure 2R) Gene symbol: PRC1, Peptide: GLAPNTPGKA (SEQ ID NO: 57). Tissues from left to right: 14 cancer tissues (1 head-and-neck cancer, 1 breast cancer, 2 esophageal cancers, 6 lung cancers, 1 ovarian cancer, 1 skin cancer, 1 urinary bladder cancer, 1 uterus cancer). Figure 3S) Gene symbol: MAPK6, Peptide: LILESIPVV (SEQ ID NO: 58). Tissues from left to right: 2 cell lines (1 blood cell, 1 skin), 25 cancer tissues (5 head-and-neck cancers, 1 colon cancer, 2 esophageal cancers, 1 leukocytic leukemia cancer, 8 lung cancers, 2 lymph node cancers, 3 skin cancers, 2 urinary bladder cancers, 1 uterus cancer). Figure 4T) Gene symbol: PPP4R1, Peptide: SLLDTLREV (SEQ ID NO: 59). Tissues from left to right: 1 normal tissue (1 small intestine), 8 cancer tissues (1 head-and-neck cancer, 2 esophageal cancers, 4 lung cancers, 1 ovarian cancer). Figure 5U) Gene symbol: TP63, Peptide: VLVPYEPPQV (SEQ ID NO: 77). Tissues from left to right: 2 normal tissues (1 esophagus, 1 trachea), 47 cancer tissues (8 head-and-neck cancers, 4 esophageal cancers, 1 gallbladder cancer, 14 lung cancers, 7 lymph node cancers, 2 prostate cancers, 1 skin cancer, 8 urinary bladder cancers. Figure 6V) Gene symbol: KIAA0947, Peptide: AVLPHVDQV (SEQ ID NO: 81). Tissues from left to right: 3 cell lines (1 blood cells, 1 pancreatic), 12 cancer tissues (5 brain cancers, 2 esophageal cancers, 1 lung cancer, 3 lymph node cancers, 1 uterus cancer).
    • Figures 2A to D show exemplary expression profiles of source genes as disclosed that are highly over-expressed or exclusively expressed in esophageal cancer in a panel of normal tissues (white bars) and 11 esophageal cancer samples (black bars). Tissues from left to right: 7 arteries, 1 brain, 1 heart, 2 livers, 2 lungs, 2 veins, 1 adipose tissue, 1 adrenal gland, 4 bone marrows, 1 colon, 2 esophagi, 2 gallbladders, 1 kidney, 6 lymph nodes, 1 pancreas, 1 pituitary gland, 1 rectum, 1 skeletal muscle, 1 skin, 1 small intestine, 1 spleen, 1 stomach, 1 thymus, 1 thyroid gland, 5 tracheae, 1 urinary bladder, 1 breast, 3 ovaries, 3 placentae, 1 prostate, 1 testis, 1 uterus, 11 esophageal cancer samples. Figure 2A) Gene symbol: PTHLH; Figure 2B) Gene symbol: KRT14; Figure 2C) Gene symbol: FAM83A; Figure 2D) Gene symbol: PDPN.
    • Figures 3A to E show exemplary results of peptide-specific in vitro CD8+ T cell responses of a healthy HLA-A*02+ donor i.e. exemplary immunogenicity data: flow cytometry results after peptide-specific multimer staining. Figure 3A) Gene symbol: SF3B3, Peptide: ELDRTPPEV (SEQ ID NO: 97); Figure 3B) Gene symbol: TNC, Peptide: AMTQLLAGV (SEQ ID NO: 101). Also, CD8+ T cells were primed using artificial APCs coated with anti-CD28 mAb and HLA-A*02 in complex with SEQ ID NO: 5 peptide (C, left panel), SEQ ID NO: 2 peptide (D, left panel) and SEQ ID NO: 77 peptide (E, left panel), respectively. After three cycles of stimulation, the detection of peptide-reactive cells was performed by 2D multimer staining with A*02/SeqID No 5 (C), A*02/SeqID No 2 (D) or A*02/SeqID No 77 (E). Right panels (C, D and E) show control staining of cells stimulated with irrelevant A*02/peptide complexes. Viable singlet cells were gated for CD8+ lymphocytes. Boolean gates helped excluding false-positive events detected with multimers specific for different peptides. Frequencies of specific multimer+ cells among CD8+ lymphocytes are indicated.
    EXAMPLES EXAMPLE 1 Identification and quantitation of tumor associated peptides presented on the cell surface Tissue samples
  • Patients' tumor tissues were obtained from Asterand (Detroit, USA and Royston, Herts, UK); ProteoGenex Inc., (Culver City, CA, USA); Tissue Solutions Ltd. (Glasgow, UK); University Hospital of Tübingen. Normal tissues were obtained from Asterand (Detroit, USA and Royston, Herts, UK); Bio-Options Inc. (CA, USA); BioServe (Beltsville, MD, USA); Capital BioScience Inc. (Rockville, MD, USA); Geneticist Inc. (Glendale, CA, USA); University Hospital of Geneva; University Hospital of Heidelberg; Kyoto Prefectural University of Medicine (KPUM); University Hospital Munich; ProteoGenex Inc. (Culver City, CA, USA); University Hospital of Tübingen; Tissue Solutions Ltd. (Glasgow, UK). Written informed consents of all patients had been given before surgery or autopsy. Tissues were shock-frozen immediately after excision and stored until isolation of TUMAPs at -70°C or below.
  • Isolation of HLA peptides from tissue samples
  • HLA peptide pools from shock-frozen tissue samples were obtained by immune precipitation from solid tissues according to a slightly modified protocol (Falk et al., 1991; Seeger et al., 1999) using the HLA-A*02-specific antibody BB7.2, the HLA-A, -B, - C-specific antibody W6/32, CNBr-activated sepharose, acid treatment, and ultrafiltration.
  • Mass spectrometry analyses
  • The HLA peptide pools as obtained were separated according to their hydrophobicity by reversed-phase chromatography (nanoAcquity UPLC system, Waters) and the eluting peptides were analyzed in LTQ- velos and fusion hybrid mass spectrometers (ThermoElectron) equipped with an ESI source. Peptide pools were loaded directly onto the analytical fused-silica micro-capillary column (75 µm i.d. x 250 mm) packed with 1.7 µm C18 reversed-phase material (Waters) applying a flow rate of 400 nL per minute. Subsequently, the peptides were separated using a two-step 180 minute-binary gradient from 10% to 33% B at a flow rate of 300 nL per minute. The gradient was composed of Solvent A (0.1% formic acid in water) and solvent B (0.1% formic acid in acetonitrile). A gold coated glass capillary (PicoTip, New Objective) was used for introduction into the nanoESI source. The LTQ-Orbitrap mass spectrometers were operated in the data-dependent mode using a TOP5 strategy. In brief, a scan cycle was initiated with a full scan of high mass accuracy in the Orbitrap (R = 30 000), which was followed by MS/MS scans also in the Orbitrap (R = 7500) on the 5 most abundant precursor ions with dynamic exclusion of previously selected ions. Tandem mass spectra were interpreted by SEQUEST and additional manual control. The identified peptide sequence was assured by comparison of the generated natural peptide fragmentation pattern with the fragmentation pattern of a synthetic sequence-identical reference peptide.
  • Label-free relative LC-MS quantitation was performed by ion counting i.e. by extraction and analysis of LC-MS features (Mueller et al., 2007). The method assumes that the peptide's LC-MS signal area correlates with its abundance in the sample. Extracted features were further processed by charge state deconvolution and retention time alignment (Mueller et al., 2008; Sturm et al., 2008). Finally, all LC-MS features were cross-referenced with the sequence identification results to combine quantitative data of different samples and tissues to peptide presentation profiles. The quantitative data were normalized in a two-tier fashion according to central tendency to account for variation within technical and biological replicates. Thus each identified peptide can be associated with quantitative data allowing relative quantification between samples and tissues. In addition, all quantitative data acquired for peptide candidates was inspected manually to assure data consistency and to verify the accuracy of the automated analysis. For each peptide a presentation profile was calculated showing the mean sample presentation as well as replicate variations. The profiles juxtapose esophageal cancer samples to a baseline of normal tissue samples.
  • Presentation profiles of exemplary over-presented peptides are shown in Figure 1. Presentation scores for exemplary peptides are shown in Table 8. Table 8: Presentation scores. The table lists peptides that are very highly over-presented on tumors compared to a panel of normal tissues (+++), highly over-presented on tumors compared to a panel of normal tissues (++) or over-presented on tumors compared to a panel of normal tissues (+).The panel of normal tissues consisted of: adipose tissue, adrenal gland, artery, vein, bone marrow, brain, central and peripheral nerve, colon, rectum, small intestine incl. duodenum, esophagus, gallbladder, heart, kidney, liver, lung, lymph node, mononuclear white blood cells, pancreas, peritoneum, pituitary, pleura, salivary gland, skeletal muscle, skin, spleen, stomach, thymus, thyroid gland, trachea, ureter, urinary bladder.
    SEQ ID No. Sequence Peptide Presentation
    1 STYGGGLSV +++
    2 SLYNLGGSKRISI +++
    3 TASAITPSV +++
    4 ALFGTILEL ++
    5 NLMASQPQL +++
    6 LLSGDLIFL +++
    7 SIFEGLLSGV +++
    8 ALLDGGSEAYWRV +++
    9 HLIAEIHTA +++
    10 SLDENSDQQV +++
    11 ALWLPTDSATV +++
    12 GLASRILDA +++
    13 SLSPVILGV +++
    14 RLPNAGTQV +++
    15 LLANGVYAA +++
    16 VLAEGGEGV +++
    17 MISRTPEV +++
    18 FLLDQVQLGL +++
    19 GLAPFLLNAV +++
    20 IIEVDPDTKEML +++
    21 IVREFLTAL +++
    22 KLNDTYVNV +++
    23 KLSDSATYL +++
    24 LLFAGTMTV +++
    25 LLPPPPPPA +++
    26 MLAEKLLQA +++
    27 NLREGDQLL +++
    28 SLDGFTIQV +++
    29 SLDGTELQL +++
    30 SLNGNQVTV +++
    32 YMLDIFHEV +++
    33 GLDVTSLRPFDL +++
    34 SLVSEQLEPA +
    35 LLRFSQDNA +++
    36 FLLRFSQDNA +++
    37 YTQPFSHYGQAL +++
    38 IAAIRGFLV +++
    39 LVRDTQSGSL +++
    40 GLAFSLYQA +++
    41 GLESEELEPEEL +
    44 ATGNDRKEAAENSL +++
    45 MLTELEKAL +++
    47 VLASGFL TV +++
    48 SMHQMLDQTL +++
    50 GMNPHQTPAQL +++
    51 KLFGHLTSA +++
    52 VAIGGVDGNVRL +++
    55 GAIDLLHNV +++
    57 GLAPNTPGKA +++
    58 LILESIPW +++
    59 SLLDTLREV +++
    61 TQTTH ELTI +++
    62 ALYEYQPLQI +++
    63 LAYTLGVKQL +++
    64 GLTDVIRDV ++
    65 YVVGGFLYQRL +++
    66 LLDEKVQSV +
    68 PAVLQSSGLYSL +++
    70 FVLDTSESV +
    71 ASDPILYRPVAV +
    72 FLPPAQVTV +
    73 KITEAIQYV +
    75 GLMDDVDFKA +
    77 VLVPYEPPQV ++
    78 KVANIIAEV +
    80 ALQEALENA ++
    81 AVLPHVDQV +++
    82 HLLGHLEQA +++
    84 SLAESLDQA +
    86 GLLTEIRAV +
    87 FLDNGPKTI +
    88 GLWEQENHL +
    89 SLADSLYNL +
    91 KLIDDVHRL +
    92 SILRHVAEV +
    94 TLLQEQGTKTV +
  • EXAMPLE 2 Expression profiling of genes encoding the peptides as disclosed
  • Over-presentation or specific presentation of a peptide on tumor cells compared to normal cells is sufficient for its usefulness in immunotherapy, and some peptides are tumor-specific despite their source protein occurring also in normal tissues. Still, mRNA expression profiling adds an additional level of safety in selection of peptide targets for immunotherapies. Especially for therapeutic options with high safety risks, such as affinity-matured TCRs, the ideal target peptide will be derived from a protein that is unique to the tumor and not found on normal tissues.
  • RNA sources and preparation
  • Surgically removed tissue specimens were provided as indicated above (see Example 1) after written informed consent had been obtained from each patient. Tumor tissue specimens were snap-frozen immediately after surgery and later homogenized with mortar and pestle under liquid nitrogen. Total RNA was prepared from these samples using TRI Reagent (Ambion, Darmstadt, Germany) followed by a cleanup with RNeasy (QIAGEN, Hilden, Germany); both methods were performed according to the manufacturer's protocol.
  • Total RNA from tumor tissue for RNASeq experiments was obtained from: ProteoGenex Inc. (Culver City, CA, USA); Tissue Solutions Ltd. (Glasgow, UK).
  • Total RNA from healthy human tissues for RNASeq experiments was obtained from: Asterand (Detroit, USA and Royston, Herts, UK); ProteoGenex Inc. (Culver City, CA, USA); Geneticist Inc. (Glendale, CA, USA); Istituto Nazionale Tumori "Pascale", Molecular Biology and Viral Oncology Unit (IRCCS) (Naples, Italy); University Hospital of Heidelberg (Germany); BioCat GmbH (Heidelberg, Germany).
  • Quality and quantity of all RNA samples were assessed on an Agilent 2100 Bioanalyzer (Agilent, Waldbronn, Germany) using the RNA 6000 Pico LabChip Kit (Agilent).
  • RNAseq experiments
  • Gene expression analysis of - tumor and normal tissue RNA samples was performed by next generation sequencing (RNAseq) by CeGaT (Tübingen, Germany). Briefly, sequencing libraries are prepared using the Illumina HiSeq v4 reagent kit according to the provider's protocol (Illumina Inc, San Diego, CA, USA), which includes RNA fragmentation, cDNA conversion and addition of sequencing adaptors. Libraries derived from multiple samples are mixed equimolarly and sequenced on the Illumina HiSeq 2500 sequencer according to the manufacturer's instructions, generating 50 bp single end reads. Processed reads are mapped to the human genome (GRCh38) using the STAR software. Expression data are provided on transcript level as RPKM (Reads Per Kilobase per Million mapped reads, generated by the software Cufflinks) and on exon level (total reads, generated by the software Bedtools), based on annotations of the ensembl sequence database (Ensembl77). Exon reads are normalized for exon length and alignment size to obtain RPKM values.
  • Exemplary expression profiles of source genes as disclosed that are highly over-expressed or exclusively expressed in esophageal cancer are shown in Figures 2. Expression scores for further exemplary genes are shown in Table 9. Table 9: Expression scores. The table lists peptides from genes that are very highly over-expressed in tumors compared to a panel of normal tissues (+++), highly over-expressed in tumors compared to a panel of normal tissues (++) or over-expressed in tumors compared to a panel of normal tissues (+). The baseline for this score was calculated from measurements of the following normal tissues: adipose tissue, adrenal gland, artery, bone marrow, brain, colon, esophagus, gallbladder, heart, kidney, liver, lung, lymph node, pancreas, pituitary, rectum, skeletal muscle, skin, small intestine, spleen, stomach, thymus, thyroid gland, trachea, urinary bladder, vein.
    SEQ ID No. Sequence Gene Expression
    1 STYGGGLSV +++
    2 SLYNLGGSKRISI +++
    3 TASAITPSV +++
    4 ALFGTILEL ++
    5 NLMASQPQL +++
    6 LLSGDLIFL +++
    7 SIFEGLLSGV +++
    8 ALLDGGSEAYWRV +++
    9 HLIAEIHTA +++
    10 SLDENSDQQV +++
    11 ALWLPTDSATV +++
    12 GLASRILDA +++
    13 SLSPVILGV +++
    14 RLPNAGTQV +++
    15 LLANGVYAA +++
    16 VLAEGGEGV +++
    17 MISRTPEV +++
    18 FLLDQVQLGL +++
    24 LLFAGTMTV +++
    25 LLPPPPPPA +
    26 MLAEKLLQA ++
    27 NLREGDQLL +++
    32 YMLDIFHEV +++
    49 GLMKDIVGA +
    55 GAIDLLHNV ++
    57 GLAPNTPGKA +
    67 SMNGGVFAV ++
    69 GLL VGSEKVTM +++
    71 ASDPILYRPVAV +
    77 VLVPYEPPQV +++
    80 ALQEALENA +
    94 TLLQEQGTKTV +++
  • EXAMPLE 3 In vitro immunogenicity for MHC class I presented peptides
  • In order to obtain information regarding the immunogenicity of the TUMAPs as disclosed, the inventors performed investigations using an in vitro T-cell priming assay based on repeated stimulations of CD8+ T cells with artificial antigen presenting cells (aAPCs) loaded with peptide/MHC complexes and anti-CD28 antibody. This way the inventors could show immunogenicity for HLA-A*0201 restricted TUMAPs of the invention, demonstrating that these peptides are T-cell epitopes against which CD8+ precursor T cells exist in humans (Table 10).
  • In vitro priming of CD8+ T cells
  • In order to perform in vitro stimulations by artificial antigen presenting cells loaded with peptide-MHC complex (pMHC) and anti-CD28 antibody, the inventors first isolated CD8+ T cells from fresh HLA-A*02 leukapheresis products via positive selection using CD8 microbeads (Miltenyi Biotec, Bergisch-Gladbach, Germany) of healthy donors obtained from the University clinics Mannheim, Germany, after informed consent.
  • PBMCs and isolated CD8+ lymphocytes were incubated in T-cell medium (TCM) until use consisting of RPMI-Glutamax (Invitrogen, Karlsruhe, Germany) supplemented with 10% heat inactivated human AB serum (PAN-Biotech, Aidenbach, Germany), 100 U/ml Penicillin/100 µg/ml Streptomycin (Cambrex, Cologne, Germany), 1 mM sodium pyruvate (CC Pro, Oberdorla, Germany), 20 µg/ml Gentamycin (Cambrex). 2.5 ng/ml IL-7 (PromoCell, Heidelberg, Germany) and 10 U/ml IL-2 (Novartis Pharma, Nürnberg, Germany) were also added to the TCM at this step.
  • Generation of pMHC/anti-CD28 coated beads, T-cell stimulations and readout was performed in a highly defined in vitro system using four different pMHC molecules per stimulation condition and 8 different pMHC molecules per readout condition.
  • The purified co-stimulatory mouse IgG2a anti human CD28 Ab 9.3 (Jung et al., 1987) was chemically biotinylated using Sulfo-N-hydroxysuccinimidobiotin as recommended by the manufacturer (Perbio, Bonn, Germany). Beads used were 5.6 µm diameter streptavidin coated polystyrene particles (Bangs Laboratories, Illinois, USA). pMHC used for positive and negative control stimulations were A*0201/MLA-001 (peptide ELAGIGILTV (SEQ ID NO. 102) from modified Melan-A/MART-1) and A*0201/DDX5-001 (YLLPAIVHI from DDX5, SEQ ID NO. 103), respectively.
  • 800.000 beads / 200 µl were coated in 96-well plates in the presence of 4 x 12.5 ng different biotin-pMHC, washed and 600 ng biotin anti-CD28 were added subsequently in a volume of 200 µl. Stimulations were initiated in 96-well plates by co-incubating 1x106 CD8+ T cells with 2x105 washed coated beads in 200 µl TCM supplemented with 5 ng/ml IL-12 (PromoCell) for 3 days at 37°C. Half of the medium was then exchanged by fresh TCM supplemented with 80 U/ml IL-2 and incubating was continued for 4 days at 37°C. This stimulation cycle was performed for a total of three times. For the pMHC multimer readout using 8 different pMHC molecules per condition, a two-dimensional combinatorial coding approach was used as previously described (Andersen et al., 2012) with minor modifications encompassing coupling to 5 different fluorochromes. Finally, multimeric analyses were performed by staining the cells with Live/dead near IR dye (Invitrogen, Karlsruhe, Germany), CD8-FITC antibody clone SK1 (BD, Heidelberg, Germany) and fluorescent pMHC multimers. For analysis, a BD LSRII SORP cytometer equipped with appropriate lasers and filters was used. Peptide specific cells were calculated as percentage of total CD8+ cells. Evaluation of multimeric analysis was done using the FlowJo software (Tree Star, Oregon, USA). In vitro priming of specific multimer+ CD8+ lymphocytes was detected by comparing to negative control stimulations. Immunogenicity for a given antigen was detected if at least one evaluable in vitro stimulated well of one healthy donor was found to contain a specific CD8+ T-cell line after in vitro stimulation (i.e. this well contained at least 1% of specific multimer+ among CD8+ T-cells and the percentage of specific multimer+ cells was at least 10x the median of the negative control stimulations).
  • In vitro immunogenicity for esophageal cancer peptides
  • For tested HLA class I peptides, in vitro immunogenicity could be demonstrated by generation of peptide specific T-cell lines. Exemplary flow cytometry results after TUMAP-specific multimer staining for two peptides (SEQ ID No 97 and SEQ ID No 101) as disclosed are shown in Figure 3 together with corresponding negative controls. Results for five peptides from the invention are summarized in Table 10A. Table 10A: in vitro immunogenicity of HLA class I peptides as disclosed Exemplary results of in vitro immunogenicity experiments conducted by the applicant for the peptides as disclosed. <20 % = +; 20 % - 49 % = ++; 50 % - 69 %= +++; >= 70 % = ++++
    SEQ ID No Sequence wells Donors
    94 TLLQEQGTKTV + ++
    95 LIQDRVAEV + ++
    97 ELDRTPPEV ++ ++++
    98 VLFPNLKTV + ++++
    101 AMTQLLAGV ++ +++
    Table 10B: In vitro immunogenicity of HLA class I peptides as disclosed Exemplary results of in vitro immunogenicity experiments conducted by the applicant for peptides as disclosed. Results of in vitro immunogenicity experiments are indicated. Percentage of positive wells and donors (among evaluable) are summarized as indicated <20 % = +; 20 % - 49 % = ++; 50 % - 69 %= +++; >= 70 %= ++++
    SEQ ID No Sequence Wells positive [%]
    1 STYGGGLSV +
    2 SLYNLGGSKRISI +
    5 NLMASQPQL +++
    6 LLSGDLIFL ++
    12 GLASRILDA +
    19 GLAPFLLNAV +
    29 SLDGTELQL +
    47 VLASGFL TV +++
    69 GLL VGSEKVTM +
    77 VLVPYEPPQV +
  • EXAMPLE 4 Synthesis of peptides
  • All peptides were synthesized using standard and well-established solid phase peptide synthesis using the Fmoc-strategy. Identity and purity of each individual peptide have been determined by mass spectrometry and analytical RP-HPLC. The peptides were obtained as white to off-white lyophilizates (trifluoro acetate salt) in purities of >50%. All TUMAPs are preferably administered as trifluoro-acetate salts or acetate salts, other salt-forms are also possible.
  • EXAMPLE 5 MHC Binding Assays
  • Candidate peptides for T cell based therapies as disclosed were further tested for their MHC binding capacity (affinity). The individual peptide-MHC complexes were produced by UV-ligand exchange, where a UV-sensitive peptide is cleaved upon UV-irradiation, and exchanged with the peptide of interest as analyzed. Only peptide candidates that can effectively bind and stabilize the peptide-receptive MHC molecules prevent dissociation of the MHC complexes. To determine the yield of the exchange reaction, an ELISA was performed based on the detection of the light chain (β2m) of stabilized MHC complexes. The assay was performed as generally described in Rodenko et al. (Rodenko et al., 2006).
  • 96 well MAXISorp plates (NUNC) were coated over night with 2ug/ml streptavidin in PBS at room temperature, washed 4x and blocked for 1h at 37°C in 2% BSA containing blocking buffer. Refolded HLA-A*02:01/MLA-001 monomers served as standards, covering the range of 15-500 ng/ml. Peptide-MHC monomers of the UV-exchange reaction were diluted 100 fold in blocking buffer. Samples were incubated for 1h at 37°C, washed four times, incubated with 2ug/ml HRP conjugated anti-β2m for 1h at 37°C, washed again and detected with TMB solution that is stopped with NH2SO4. Absorption was measured at 450nm. Candidate peptides that show a high exchange yield (preferably higher than 50%, most preferred higher than 75%) are generally preferred for a generation and production of antibodies or fragments thereof, and/or T cell receptors or fragments thereof, as they show sufficient avidity to the MHC molecules and prevent dissociation of the MHC complexes. Table 11: MHC class I binding scores.
    Binding of HLA-class I restricted peptides to HLA-A*02:01 was ranged by peptide exchange yield: ≥10% = +; ≥20% = ++; ≥50 = +++; ≥ 75% = ++++
    SEQ ID Sequence Peptide exchange
    1 STYGGGLSV +++
    2 SLYNLGGSKRISI ++++
    3 TASAITPSV +++
    5 NLMASQPQL +++
    6 LLSGDLIFL +++
    7 SIFEGLLSGV ++
    8 ALLDGGSEAYWRV +++
    9 HLIAEIHTA +++
    10 SLDENSDQQV +++
    11 ALWLPTDSATV +++
    12 GLASRILDA +++
    13 SLSPVILGV ++++
    14 RLPNAGTQV ++++
    15 LLANGVYAA +
    16 VLAEGGEGV +++
    17 MISRTPEV ++
    18 FLLDQVQLGL +++
    19 GLAPFLLNAV +++
    21 IVREFLTAL +++
    22 KLNDTYVNV +++
    23 KLSDSATYL +++
    24 LLFAGTMTV ++
    25 LLPPPPPPA +++
    26 MLAEKLLQA +
    27 NLREGDQLL +++
    28 SLDGFTIQV ++
    29 SLDGTELQL +++
    30 SLNGNQVTV +
    31 VLPKLYVKL ++
    32 YMLDIFHEV ++
    33 GLDVTSLRPFDL +++
    34 SLVSEQLEPA +++
    35 LLRFSQDNA +++
    36 FLLRFSQDNA ++
    37 YTQPFSHYGQAL +++
    38 IAAIRGFLV +++
    SEQ ID Sequence Peptide exchange
    39 LVRDTQSGSL ++
    40 GLAFSLYQA ++
    41 GLESEELEPEEL ++
    42 TQTAVITRI +
    43 KVVGKDYLL +
    44 ATGNDRKEAAENSL +++
    45 MLTELEKAL ++
    46 YTAQIGADIAL +++
    47 VLASGFL TV ++++
    48 SMHQMLDQTL ++
    49 GLMKDIVGA +++
    51 KLFGHLTSA ++
    52 VAIGGVDGNVRL ++
    53 WVTGLTLV ++
    54 YQDLLNVKM +++
    55 GAIDLLHNV ++
    56 ALVEVTEHV ++
    57 GLAPNTPGKA +++
    58 LILESIPVV ++
    59 SLLDTLREV +++
    60 WMEELLKV ++
    61 TQTTH ELTI +++
    62 ALYEYQPLQI ++
    63 LAYTLGVKQL +++
    64 GLTDVIRDV ++++
    65 YVVGGFLYQRL +++
    66 LLDEKVQSV +++
    67 SMNGGVFAV ++
    68 PAVLQSSGLYSL ++
    69 GLL VGSEKVTM +++
    70 FVLDTSESV +++
    71 ASDPILYRPVAV +++
    72 FLPPAQVTV ++
    73 KITEAIQYV +++
    74 ILASLATSV +++
    76 KVADYIPQL +++
    77 VLVPYEPPQV ++
    78 KVANIIAEV ++
    79 GQDVGRYQV ++
    80 ALQEALENA ++
    81 AVLPHVDQV +++
    82 HLLGHLEQA +++
    83 ALADGVVSQA +++
    84 SLAESLDQA +++
    85 NIIELVHQV ++++
    87 FLDNGPKTI +++
    89 SLADSLYNL ++
    90 SIYEYYHAL +++
    91 KLIDDVHRL ++++
    92 SILRHVAEV ++
    93 VLINTSVTL +++
  • EXAMPLE 6 Absolute quantitation of tumor associated peptides presented on the cell surface
  • The generation of binders, such as antibodies and/or TCRs, is a laborious process, which may be conducted only for a number of selected targets. In the case of tumor-associated and -specific peptides, selection criteria include but are not restricted to exclusiveness of presentation and the density of peptide presented on the cell surface. The quantitation of TUMAP copies per cell in solid tumor samples requires the absolute quantitation of the isolated TUMAP, the efficiency of TUMAP isolation, and the cell count of the tissue sample analyzed.
  • Peptide quantitation by nanoLC-MS/MS
  • For an accurate quantitation of peptides by mass spectrometry, a calibration curve was generated for each peptide using the internal standard method. The internal standard is a double-isotope-labelled variant of each peptide, i.e. two isotope-labelled amino acids were included in TUMAP synthesis. It differs from the tumor-associated peptide only in its mass but shows no difference in other physicochemical properties (Anderson et al., 2012). The internal standard was spiked to each MS sample and all MS signals were normalized to the MS signal of the internal standard to level out potential technical variances between MS experiments.
  • The calibration curves were prepared in at least three different matrices, i.e. HLA peptide eluates from natural samples similar to the routine MS samples, and each preparation was measured in duplicate MS runs. For evaluation, MS signals were normalized to the signal of the internal standard and a calibration curve was calculated by logistic regression.
  • For the quantitation of tumor-associated peptides from tissue samples, the respective samples were also spiked with the internal standard; the MS signals were normalized to the internal standard and quantified using the peptide calibration curve.
  • Efficiency of peptide/MHC isolation
  • As for any protein purification process, the isolation of proteins from tissue samples is associated with a certain loss of the protein of interest. To determine the efficiency of TUMAP isolation, peptide/MHC complexes were generated for all TUMAPs selected for absolute quantitation. To be able to discriminate the spiked from the natural peptide/MHC complexes, single-isotope-labelled versions of the TUMAPs were used, i.e. one isotope-labelled amino acid was included in TUMAP synthesis. These complexes were spiked into the freshly prepared tissue lysates, i.e. at the earliest possible point of the TUMAP isolation procedure, and then captured like the natural peptide/MHC complexes in the following affinity purification. Measuring the recovery of the single-labelled TUMAPs therefore allows conclusions regarding the efficiency of isolation of individual natural TUMAPs.
  • The efficiency of isolation was analyzed in a low number of samples and was comparable among these tissue samples. In contrast, the isolation efficiency differs between individual peptides. This suggests that the isolation efficiency, although determined in only a limited number of tissue samples, may be extrapolated to any other tissue preparation. However, it is necessary to analyze each TUMAP individually as the isolation efficiency may not be extrapolated from one peptide to others.
  • Determination of the cell count in solid, frozen tissue
  • In order to determine the cell count of the tissue samples subjected to absolute peptide quantitation, the inventors applied DNA content analysis. This method is applicable to a wide range of samples of different origin and, most importantly, frozen samples (Alcoser et al., 2011; Forsey and Chaudhuri, 2009; Silva et al., 2013). During the peptide isolation protocol, a tissue sample is processed to a homogenous lysate, from which a small lysate aliquot is taken. The aliquot is divided in three parts, from which DNA is isolated (QiaAmp DNA Mini Kit, Qiagen, Hilden, Germany). The total DNA content from each DNA isolation is quantified using a fluorescence-based DNA quantitation assay (Qubit dsDNA HS Assay Kit, Life Technologies, Darmstadt, Germany) in at least two replicates.
  • In order to calculate the cell number, a DNA standard curve from aliquots of single healthy blood cells, with a range of defined cell numbers, has been generated. The standard curve is used to calculate the total cell content from the total DNA content from each DNA isolation. The mean total cell count of the tissue sample used for peptide isolation is extrapolated considering the known volume of the lysate aliquots and the total lysate volume.
  • Peptide copies per cell
  • With data of the aforementioned experiments, the inventors calculated the number of TUMAP copies per cell by dividing the total peptide amount by the total cell count of the sample, followed by division through isolation efficiency. Copy cell numbers for selected peptides are shown in Table 12. Table 12: Absolute copy numbers. The table lists the results of absolute peptide quantitation in NSCLC tumor samples. The median number of copies per cell are indicated for each peptide: <100 = +; >=100 = ++; >=1,000 +++; >=10,000 = ++++. The number of samples, in which evaluable, high quality MS data are available, is indicated.
    SEQ ID No. Peptide Code Copies per cell (median) Number of samples
    9 PTHL-001 + 31
  • Reference List
    • Abbas, W. et al., Front Oncol 5 (2015): 75
    • Adams, S. et al., PLoS.One. 9 (2014): e112945
    • Al Moustafa, A. E. et al., Oncogene 21 (2002): 2634-2640
    • Al-Mahdi, R. et al., Cell Adh.Migr. (2015): 0
    • Alcoser, S. Y. et al., BMC.Biotechnol. 11 (2011): 124
    • Alholle, A. et al., Epigenetics. 8 (2013): 1198-1204
    • Ali, R. H. et al., Hum.Pathol. 45 (2014): 2453-2462
    • Allison, J. P. et al., Science 270 (1995): 932-933
    • Alper, M. et al., Mol.Cell Biochem. 393 (2014): 165-175
    • Alsagaby, S. A. et al., J Proteome.Res 13 (2014): 5051-5062
    • Altmannsberger, M. et al., Am.J Pathol. 118 (1985): 85-95
    • Ammendola, M. et al., PLoS.One. 9 (2014): e99512
    • Andersen, R. S. et al., Nat.Protoc. 7 (2012): 891-902
    • Anderson, N. L. et al., J Proteome.Res 11 (2012): 1868-1878
    • Appay, V. et al., Eur.J Immunol. 36 (2006): 1805-1814
    • Arentz, G. et al., Clin Proteomics. 8 (2011): 16
    • Arif, Q. et al., Arch.Pathol.Lab Med. 139 (2015): 978-980
    • Auvinen, P. et al., Breast Cancer Res Treat. 143 (2014): 277-286
    • Avasarala, S. et al., PLoS.One. 8 (2013): e76895
    • Banchereau, J. et al., Cell 106 (2001): 271-274
    • Bandres, E. et al., Oncol Rep. 12 (2004): 287-292
    • Banerjee, K. et al., Int.J Cancer (2015)
    • Barros-Filho, M. C. et al., J Clin Endocrinol.Metab 100 (2015): E890-E899
    • Bashyam, M. D. et al., Neoplasia. 7 (2005): 556-562
    • Basu, S. et al., PLoS.One. 10 (2015): e0123979
    • Baxter, P. A. et al., Acta Neuropathol.Commun. 2 (2014): 160
    • Beatty, G. et al., J Immunol 166 (2001): 2276-2282
    • Becker, S. A. et al., Cancer Res 56 (1996): 5092-5097
    • Beggs, J. D., Nature 275 (1978): 104-109
    • Bellon, M. et al., Blood 121 (2013): 5045-5054
    • Benjamini, Y. et al., Journal of the Royal Statistical Society.Series B (Methodological), Vol.57 (1995): 289-300
    • Bhattacharjee, R. B. et al., Cell Biol Int. 36 (2012): 697-704
    • Bin Amer, S. M. et al., Saudi.Med.J 29 (2008): 507-513
    • Blanch, A. et al., PLoS.One. 8 (2013): e66436
    • Blanco, M. A. et al., Cell Res 22 (2012): 1339-1355
    • Blenk, S. et al., Cancer Inform. 3 (2007): 399-420
    • Boulter, J. M. et al., Protein Eng 16 (2003): 707-711
    • Boyer, A. P. et al., Mol.Cell Proteomics. 12 (2013): 180-193
    • Bozza, W. P. et al., Oncotarget. 6 (2015): 32723-32736
    • Braulke, T. et al., Arch.Biochem.Biophys. 298 (1992): 176-181
    • Braumuller, H. et al., Nature (2013)
    • Bray, F. et al., Int J Cancer 132 (2013): 1133-1145
    • Brechmann, M. et al., Immunity. 37 (2012): 697-708
    • Bredholt, G. et al., Oncotarget. 6 (2015): 39676-39691
    • Breuninger, S. et al., Am.J Pathol. 176 (2010): 2509-2519
    • Brezinova, J. et al., Cancer Genet.Cytogenet. 173 (2007): 10-16
    • Broghammer, M. et al., Cancer Lett. 214 (2004): 225-229
    • Brossart, P. et al., Blood 90 (1997): 1594-1599
    • Bruckdorfer, T. et al., Curr.Pharm.Biotechnol. 5 (2004): 29-43
    • Buckley, N. E. et al., Cell Death.Dis. 5 (2014): e1070
    • Bui, P. H. et al., Mol.Pharmacol. 76 (2009): 1044-1052
    • Buim, M. E. et al., Oncology 69 (2005): 445-454
    • Bujas, T. et al., Eur.J Histochem. 55 (2011): e7
    • Cai, J. L. et al., Chin J Cancer Res 23 (2011): 59-63
    • Cai, Q. et al., Nat Genet. 46 (2014): 886-890
    • Calmon, M. F. et al., Epigenetics. 10 (2015): 622-632
    • Calvo, N. et al., Biochem.Cell Biol 92 (2014): 305-315
    • Canet, B. et al., Hum.Pathol. 42 (2011): 833-839
    • Cao, Z. et al., Mol.Oncol 8 (2014): 285-296
    • Card, K. F. et al., Cancer Immunol Immunother. 53 (2004): 345-357
    • Carinci, F. et al., Int.J Immunopathol.Pharmacol. 18 (2005): 513-524
    • Cazier, J. B. et al., Nat Commun. 5 (2014): 3756
    • Cetindis, M. et al., Eur.Arch.Otorhinolaryngol. (2015)
    • Chakrabarti, G. et al., Cancer Metab 3 (2015): 12
    • Chaneton, B. et al., Trends Biochem.Sci. 37 (2012): 309-316
    • Chang, I. W. et al., Tumour.Biol 36 (2015): 5441-5450
    • Chang, J. W. et al., Anticancer Res 32 (2012): 1259-1265
    • Chanock, S. J. et al., Hum.Immunol. 65 (2004): 1211-1223
    • Chauvet, C. et al., PLoS.One. 6 (2011): e22545
    • Che, J. et al., Tumour.Biol 36 (2015): 6559-6568
    • Chen, B. et al., Mol.Cancer Res 10 (2012): 305-315
    • Chen, K. D. et al., Cell Death.Dis. 5 (2014a): e1244
    • Chen, L. et al., Int.J Mol.Sci. 15 (2014b): 11435-11445
    • Chen, Q. et al., Cell Physiol Biochem. 35 (2015a): 1052-1061
    • Chen, R. S. et al., Oncogene 28 (2009): 599-609
    • Chen, S. et al., Cancer Epidemiol. 37 (2013): 172-178
    • Chen, W. M. et al., Dig.Dis.Sci. 60 (2015b): 1655-1662
    • Chen, Y. et al., Med.Oncol 31 (2014c): 304
    • Chen, Y. C. et al., Int.J Cancer 135 (2014d): 117-127
    • Chen, Z. T. et al., Int.J Mol.Sci. 16 (2015c): 15497-15530
    • Cheon, D. J. et al., Clin Cancer Res 20 (2014): 711-723
    • Cheuk, W. et al., Pathology 33 (2001): 7-12
    • Cho, S. J. et al., PLoS.One. 8 (2013): e71724
    • Choi, W. I. et al., Cell Physiol Biochem. 23 (2009): 359-370
    • Chuang, W. Y. et al., Histol.Histopathol. 28 (2013): 293-299
    • Chung, J. et al., J Cell Biol 158 (2002): 165-174
    • Chung, T. K. et al., Int.J Cancer 137 (2015): 776-783
    • Cimino, D. et al., Int.J Cancer 123 (2008): 1327-1338
    • Cipriano, R. et al., Mol.Cancer Res 12 (2014): 1156-1165
    • ClinicalTrials.gov, (2015), http://www.clinicaltrials.gov
    • Cohen, C. J. et al., J Mol Recognit. 16 (2003a): 324-332
    • Cohen, C. J. et al., J Immunol 170 (2003b): 4349-4361
    • Cohen, S. N. et al., Proc.Natl.Acad.Sci.U.S.A 69 (1972): 2110-2114
    • Coligan, J. E. et al., Current Protocols in Protein Science (1995)
    • Colombetti, S. et al., J Immunol. 176 (2006): 2730-2738
    • Council, L. et al., Mod.Pathol. 22 (2009): 639-650
    • Crossen, P. E. et al., Cancer Genet.Cytogenet. 113 (1999): 126-133
    • Cui, D. et al., Oncogene 33 (2014): 2225-2235
    • Daigeler, A. et al., J Exp.Clin Cancer Res 27 (2008): 82
    • Dang, C. V. et al., Clin Cancer Res 15 (2009): 6479-6483
    • Dang, Q. et al., Med.Oncol 31 (2014): 24
    • Dar, A. A. et al., Immunology (2015)
    • Davids, M. S. et al., Leuk.Lymphoma 53 (2012): 2362-2370
    • Davidson, B. et al., Hum.Pathol. 45 (2014): 691-700
    • de Groen, F. L. et al., Genes Chromosomes.Cancer 53 (2014): 339-348
    • de Jonge, H. J. et al., Leukemia 25 (2011): 1825-1833
    • de Sa, V. K. et al., Braz.J Med.Biol Res 46 (2013): 21-31
    • De, Keersmaecker K. et al., Haematologica 99 (2014): 85-93
    • De, Ponti A. et al., Cancer Lett. 369 (2015): 396-404
    • Deacu, E. et al., Cancer Res 64 (2004): 7690-7696
    • Deb, S. et al., Mod.Pathol. 27 (2014): 1223-1230
    • Debiec-Rychter, M. et al., Genes Chromosomes.Cancer 38 (2003): 187-190
    • DeLaBarre, B. et al., Chem Biol 21 (2014): 1143-1161
    • Delker, D. A. et al., PLoS.One. 9 (2014): e88367
    • Demirag, G. G. et al., Med.Oncol 29 (2012): 1518-1522
    • Demirci, H. et al., J Ophthalmic Vis.Res 8 (2013): 303-307
    • Dengjel, J. et al., Clin Cancer Res 12 (2006): 4163-4170
    • Denkberg, G. et al., J Immunol 171 (2003): 2197-2207
    • Depianto, D. et al., Nat Genet. 42 (2010): 910-914
    • Ding, L. et al., Nature 481 (2012): 506-510
    • Dotlic, S. et al., Appl.Immunohistochem.Mol.Morphol. 22 (2014): 537-542
    • Downie, D. et al., Clin Cancer Res. 11 (2005): 7369-7375
    • Draberova, E. et al., J Neuropathol.Exp.Neurol. 74 (2015): 723-742
    • Drayton, R. M. et al., Clin Cancer Res 20 (2014): 1990-2000
    • Du, J. et al., Int.J Mol.Sci. 13 (2012): 15755-15766
    • Duanmin, H. et al., Hepatogastroenterology 60 (2013): 870-875
    • Dubrowinskaja, N. et al., Cancer Med. 3 (2014): 300-309
    • Ehrlichova, M. et al., Genomics 102 (2013): 96-101
    • El-Naggar, A. M. et al., Cancer Cell 27 (2015): 682-697
    • Espinosa, A. M. et al., PLoS.One. 8 (2013): e55975
    • Esseghir, S. et al., J Pathol. 210 (2006): 420-430
    • Falk, K. et al., Nature 351 (1991): 290-296
    • Fang, J. et al., BMC.Cancer 8 (2008a): 69
    • Fang, W. et al., J Transl.Med. 6 (2008b): 32
    • Fejzo, M. S. et al., Int.J Mol.Sci. 14 (2013): 3094-3109
    • Fellenberg, F. et al., J Invest Dermatol. 122 (2004): 1510-1517
    • Feng, G. et al., Leuk.Lymphoma 55 (2014): 2699-2705
    • Ferlay et al., GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No.11 [Internet], (2013), http://qlobocan.iarc.fr
    • Fong, L. et al., Proc.Natl.Acad.Sci.U.S.A 98 (2001): 8809-8814
    • Fontaine, J. F. et al., PLoS.One. 4 (2009): e7632
    • Forsey, R. W. et al., Biotechnol.Lett. 31 (2009): 819-823
    • Fu, A. et al., Mol.Med.Rep. 11 (2015a): 4727-4733
    • Fu, Y. et al., Cancer Biol.Ther 5 (2006): 741-744
    • Fu, Z. C. et al., Med.Sci.Monit. 21 (2015b): 1276-1287
    • Fujita, A. et al., Genet.Mol.Res 7 (2008): 371-378
    • Fujita, N. et al., J Biochem. 152 (2012): 407-413
    • Fujiwara, K. et al., PLoS.One. 9 (2014): e107247
    • Furukawa, C. et al., Cancer Res 65 (2005): 7102-7110
    • Gabrilovich, D. I. et al., Nat Med. 2 (1996): 1096-1103
    • Gao, Y. B. et al., Nat Genet. 46 (2014): 1097-1102
    • Gattinoni, L. et al., Nat Rev.lmmunol 6 (2006): 383-393
    • Giallourakis, C. C. et al., J Immunol. 190 (2013): 5578-5587
    • Giovinazzo, F. et al., Cell Signal. 25 (2013): 651-659
    • Gkika, D. et al., J Cell Biol 208 (2015): 89-107
    • Gnjatic, S. et al., Proc Natl.Acad.Sci.U.S.A 100 (2003): 8862-8867
    • Godkin, A. et al., Int.Immunol 9 (1997): 905-911
    • Goode, G. et al., PLoS.One. 9 (2014): e100103
    • Gordon, G. J. et al., BMC.Cancer 11 (2011): 169
    • Gorski, J. J. et al., Breast Cancer Res Treat. 122 (2010): 721-731
    • Green, M. R. et al., Molecular Cloning, A Laboratory Manual 4th (2012)
    • Greenfield, E. A., Antibodies: A Laboratory Manual 2nd (2014)
    • Groulx, J. F. et al., Carcinogenesis 35 (2014): 1217-1227
    • Guo, X. et al., Sci.Rep. 5 (2015): 11846
    • Gupta, V. et al., Curr.Pharm.Des 20 (2014): 2595-2606
    • Hagel, C. et al., J Neurooncol. 112 (2013): 191-197
    • Haggman, M. J. et al., Urology 50 (1997): 643-647
    • Hammam, O. et al., J Egypt.Soc.Parasitol. 44 (2014): 733-740
    • Hapgood, G. et al., Blood 126 (2015): 17-25
    • Haraguchi, N. et al., Int.J Oncol 43 (2013): 425-430
    • Harris, T. M. et al., Arch.Pathol.Lab Med. 139 (2015): 494-507
    • Haslene-Hox, H. et al., Biochim.Biophys.Acta 1834 (2013): 2347-2359
    • Hatina, J. et al., Neoplasma 59 (2012): 728-736
    • Hauser, A. D. et al., Mol.Cancer Res 12 (2014): 130-142
    • He, X. et al., Int.J Biol Macromol. 72 (2015): 1081-1089
    • Heffler, M. et al., Anticancer Agents Med.Chem 13 (2013): 584-594
    • Heubeck, B. et al., Eur.J Cancer 49 (2013): e1-e7
    • Hoeflich, K. P. et al., Int.J Oncol 29 (2006): 839-849
    • Hofsli, E. et al., Br.J Cancer 99 (2008): 1330-1339
    • Hogan, L. E. et al., Blood 118 (2011): 5218-5226
    • Hountis, P. et al., Tumour.Biol 35 (2014): 7327-7333
    • Hu, C. K. et al., Mol.Biol Cell 23 (2012): 2702-2711
    • Huang, F. et al., Int.J Clin Exp.Pathol. 7 (2014a): 1093-1100
    • Huang, S. L. et al., Cancers (Basel) 7 (2015): 1052-1071
    • Huang, Y. D. et al., Hua Xi.Kou Qiang.Yi.Xue.Za Zhi. 25 (2007): 500-503
    • Huang, Z. et al., Indian J Otolaryngol.Head Neck Surg. 66 (2014b): 120-125
    • Huang, Z. et al., J Oral Pathol.Med. 43 (2014c): 191-198
    • Hussey, G. S. et al., Mol Cell 41 (2011): 419-431
    • Hwang, M. L. et al., J Immunol. 179 (2007): 5829-5838
    • Ichinose, J. et al., Cancer Sci. 105 (2014): 1135-1141
    • Ida-Yonemochi, H. et al., Mod.Pathol. 25 (2012): 784-794
    • Ii, M. et al., Int.J Oncol 39 (2011): 593-599
    • Inoue, H. et al., Int.J Cancer 63 (1995): 523-526
    • Inoue, K. et al., Subcell.Biochem. 85 (2014): 17-40
    • Iqbal, M. A. et al., FEBS Lett. 588 (2014): 2685-2692
    • Irifune, H. et al., Cancer Biol Ther. 4 (2005): 449-455
    • Ismail, M. F. et al., Tumour.Biol (2015)
    • Israelsen, W. J. et al., Semin.Cell Dev.Biol 43 (2015): 43-51
    • Jamieson, N. B. et al., Clin Cancer Res 17 (2011): 3316-3331
    • Januchowski, R. et al., Biomed.Pharmacother. 67 (2013): 240-245
    • Jiang, L. et al., Cell Cycle 14 (2015): 2881-2885
    • Jin, J. et al., Int.J Hematol. 99 (2014): 750-757
    • Johnson, R. H. et al., Oncotarget. (2015)
    • Johnstone, C. N. et al., Dis.Model.Mech. 8 (2015): 237-251
    • Joosse, S. A. et al., Clin Cancer Res 18 (2012): 993-1003
    • Jung, G. et al., Proc Natl Acad Sci U S A 84 (1987): 4611-4615
    • Kabbage, M. et al., J Biomed.Biotechnol. 2008 (2008): 564127
    • Kanehira, M. et al., Cancer Res 67 (2007): 3276-3285
    • Kang, S. et al., J Proteome.Res 9 (2010): 5638-5645
    • Kao, C. J. et al., Oncogene 27 (2008): 1397-1403
    • Karlsson, J. et al., Cancer Lett. 357 (2015): 498-501
    • Kato, I. et al., Pathol.Int. 59 (2009): 38-43
    • Katoh, M., Int.J Oncol 41 (2012): 1913-1918
    • Kaz, A. M. et al., Genes Chromosomes.Cancer 51 (2012): 384-393
    • Kazma, R. et al., PLoS.One. 7 (2012): e51680
    • Kerley-Hamilton, J. S. et al., Oncogene 24 (2005): 6090-6100
    • Khakpour, G. et al., Tumour.Biol 36 (2015): 4905-4912
    • Khalil, A. A., Cancer Sci. 98 (2007): 201-213
    • Khapare, N. et al., PLoS.One. 7 (2012): e38561
    • Kibbe, A. H., Handbook of Pharmaceutical Excipients rd (2000)
    • Kido, T. et al., Genes (Basel) 1 (2010): 283-293
    • Kim, S. W. et al., OMICS. 15 (2011): 281-292
    • Kimura, H. et al., Int.J Oncol 30 (2007): 171-179
    • Kinoshita, T. et al., Oncotarget. 3 (2012): 1386-1400
    • Kirov, A. et al., J Cell Biochem. (2015)
    • Kita, Y. et al., Eur.J Surg.Oncol 35 (2009): 52-58
    • Klopfleisch, R. et al., J Proteome.Res 9 (2010): 6380-6391
    • Koizume, S. et al., Cancer Res 66 (2006): 9453-9460
    • Koizume, S. et al., World J Clin Oncol 5 (2014): 908-920
    • Koizume, S. et al., Biomark.Cancer 7 (2015): 1-13
    • Kono, K. et al., Cancer Sci. 100 (2009): 1502-1509
    • Kottorou, A. E. et al., Acta Histochem. 114 (2012): 553-561
    • Krieg, A. M., Nat Rev.Drug Discov. 5 (2006): 471-484
    • Krisenko, M. O. et al., Biochim.Biophys.Acta 1853 (2015): 254-263
    • Kruse, A. J. et al., Int.J Gynecol.Cancer 24 (2014): 1616-1622
    • Kulkarni, G. et al., Breast Cancer Res Treat. 102 (2007): 31-41
    • Kultti, A. et al., Biomed.Res Int. 2014 (2014): 817613
    • Kumar, M. et al., J Transl.Med. 13 (2015): 8
    • Kumarakulasingham, M. et al., Clin Cancer Res 11 (2005): 3758-3765
    • Kuramitsu, Y. et al., Anticancer Res 30 (2010): 4459-4465
    • Kurimoto, F. et al., Int.J Mol.Med. 8 (2001): 89-93
    • Kwon, O. H. et al., Biochem.Biophys.Res Commun. 406 (2011): 539-545
    • Larrinaga, G. et al., Dis.Markers 35 (2013): 825-832
    • Lauvrak, S. U. et al., Br.J Cancer 109 (2013): 2228-2236
    • Leal, M. F. et al., World J Gastroenterol. 18 (2012): 1531-1537
    • Ledet, E. M. et al., Prostate 73 (2013): 614-623
    • Lee, E. J. et al., J Genet.Genomics 42 (2015a): 355-371
    • Lee, H. W. et al., Dis.Esophagus. (2015b)
    • Lee, J. et al., Oncoscience. 2 (2015c): 410-418
    • Lee, J. M., Reprod.Biol Endocrinol. 1 (2003): 69
    • Lee, M. H. et al., J Cell Sci. 126 (2013): 1744-1752
    • Lee, M. H. et al., Ann.N.Y.Acad.Sci. 1171 (2009): 87-93
    • Lee, S. H. et al., EMBO J 25 (2006): 4008-4019
    • Lee, S. Y. et al., J Clin Invest 122 (2012): 3211-3220
    • Lei, Y. Y. et al., Asian Pac.J Cancer Prev. 15 (2014): 8539-8548
    • Leithner, K. et al., BMC.Cancer 14 (2014): 40
    • Leitlinie Magenkarzinom, 032-009OL, (2012)
    • Lexander, H. et al., Anal.Quant.Cytol.Histol. 27 (2005): 263-272
    • Li, C. et al., Oncogene 23 (2004): 9336-9347
    • Li, C. et al., Am.J Cancer Res 5 (2015a): 1635-1648
    • Li, R. et al., Oncogene 25 (2006): 2628-2635
    • Li, X. et al., Pancreas 40 (2011): 753-761
    • Li, X. Q. et al., PLoS.One. 7 (2012): e31146
    • Li, Y. et al., Neoplasia. 7 (2005): 1073-1080
    • Li, Y. et al., Cell Rep. 12 (2015b): 388-395
    • Li, Y. et al., Lung Cancer 58 (2007): 171-183
    • Li, Z. et al., Biochim.Biophys.Acta 1846 (2014): 285-296
    • Liang, B. et al., World J Gastroenterol. 11 (2005a): 623-628
    • Liang, J. et al., Tumour.Biol 36 (2015): 6391-6399
    • Liang, L. et al., Int.J Oncol 45 (2014): 659-666
    • Liang, Z. et al., Zhonghua Zhong.Liu Za Zhi. 27 (2005b): 534-537
    • Liao, J. S. et al., Zhejiang.Da.Xue.Xue.Bao.Yi.Xue.Ban. 44 (2015a): 329-334
    • Liao, W. et al., Oncotarget. 6 (2015b): 24132-24147
    • Liddy, N. et al., Nat Med. 18 (2012): 980-987
    • Lim, M. Y. et al., Int.J Chronic.Dis. 2013 (2013): 578613
    • Lim, R. et al., Biochem.Biophys.Res Commun. 406 (2011): 408-413
    • Lin, C. et al., Oncotarget. 6 (2015): 8434-8453
    • Lin, S. J. et al., J Proteomics. 94 (2013): 186-201
    • Lin, X. et al., Med.Oncol 31 (2014): 42
    • Linher-Melville, K. et al., Mol.Cell Biochem. 405 (2015): 205-221
    • Liu, B. et al., J Clin Endocrinol.Metab 99 (2014a): E786-E795
    • Liu, C. et al., Int.J Clin Exp.Pathol. 7 (2014b): 690-698
    • Liu, D. et al., Genet.Mol.Res 13 (2014c): 8153-8162
    • Liu, J. P. et al., Zhonghua Yi.Xue.Za Zhi. 87 (2007a): 2719-2723
    • Liu, L. et al., Oncol Lett. 7 (2014d): 2192-2198
    • Liu, Y. et al., J Neurooncol. 99 (2010): 13-24
    • Liu, Y. et al., Cell Death.Dis. 6 (2015): e1630
    • Liu, Y. et al., Oncol Rep. 18 (2007b): 943-951
    • Liu, Y. F. et al., Tumour.Biol 35 (2014e): 3731-3741
    • Ljunggren, H. G. et al., J Exp.Med. 162 (1985): 1745-1759
    • Lo, P. K. et al., Oncogene 31 (2012): 2614-2626
    • Lo, R. K. et al., J Biol Chem 278 (2003): 52154-52165
    • Lo, W. Y. et al., J Proteome.Res 6 (2007): 2143-2151
    • Lohr, J. G. et al., Cancer Cell 25 (2014): 91-101
    • Long, W. et al., J Clin Invest 122 (2012): 1869-1880
    • Longenecker, B. M. et al., Ann N.Y.Acad.Sci. 690 (1993): 276-291
    • Longerich, T., Pathologe 35 Suppl 2 (2014): 177-184
    • Lonsdale, J., Nat.Genet. 45 (2013): 580-585
    • Lou, S. et al., Stem Cells 31 (2013): 1942-1953
    • Lu, Z. et al., Cell Physiol Biochem. 33 (2014): 859-868
    • Lukas, T. J. et al., Proc.Natl.Acad.Sci.U.S.A 78 (1981): 2791-2795
    • Lundblad, R. L., Chemical Reagents for Protein Modification 3rd (2004)
    • Luo, W. et al., Trends Endocrinol.Metab 23 (2012): 560-566
    • Lv, Z. et al., J Exp.Clin Cancer Res 33 (2014): 100
    • Ma, Y. et al., Mol.Cell Proteomics. 8 (2009): 1878-1890
    • Madanayake, T. W. et al., BMC.Genomics 14 (2013): 833
    • Maiso, P. et al., Cancer Res 75 (2015): 2071-2082
    • Manenti, G. et al., Toxicol.Lett. 112-113 (2000): 257-263
    • Mao, P. et al., J Biol Chem 286 (2011): 19381-19391
    • Mao, P. et al., PLoS.One. 8 (2013): e81803
    • Marcinkiewicz, K. M. et al., Exp.Cell Res 320 (2014): 128-143
    • Marg, A. et al., Biochem.Biophys.Res Commun. 401 (2010): 143-148
    • Marhold, M. et al., Mol.Cancer Res 13 (2015): 556-564
    • Martin-Rufian, M. et al., J Mol.Med.(Berl) 92 (2014): 277-290
    • Matassa, D. S. et al., Cell Death.Dis. 4 (2013): e851
    • Mayas, M. D. et al., Anticancer Res 32 (2012): 3675-3682
    • Mazan-Mamczarz, K. et al., PLoS.Genet. 10 (2014): e1004105
    • Mazurek, S., Ernst.Schering.Found.Symp.Proc. (2007): 99-124
    • Mazurek, S., Int.J Biochem.Cell Biol 43 (2011): 969-980
    • McBride, D. J. et al., J Pathol. 227 (2012): 446-455
    • Melaiu, O. et al., Mutat.Res 771 (2015): 6-12
    • Messina, M. et al., Blood 123 (2014): 2378-2388
    • Meziere, C. et al., J Immunol 159 (1997): 3230-3237
    • Mimori, K. et al., Int.J Oncol 11 (1997): 959-964
    • Mirza, Z. et al., Anticancer Res 34 (2014): 1873-1884
    • Missero, C. et al., Exp.Dermatol. 23 (2014): 143-146
    • Moretti, R. M. et al., Oncol Rep. 9 (2002): 1139-1143
    • Morgan, R. A. et al., Science 314 (2006): 126-129
    • Mori, M. et al., Transplantation 64 (1997): 1017-1027
    • Morris, L. G. et al., Nat Genet. 45 (2013): 253-261
    • Mortara, L. et al., Clin Cancer Res. 12 (2006): 3435-3443
    • Mountzios, G. et al., Ann.Oncol 25 (2014): 1889-1900
    • Mueller, L. N. et al., J Proteome.Res 7 (2008): 51-61
    • Mueller, L. N. et al., Proteomics. 7 (2007): 3470-3480
    • Mumberg, D. et al., Proc.Natl.Acad.Sci.U.S.A 96 (1999): 8633-8638
    • Murray, G. I. et al., Histopathology 57 (2010): 202-211
    • Mustacchi, G. et al., Int.J Mol.Sci. 14 (2013): 9686-9702
    • Naba, A. et al., Elife. 3 (2014): e01308
    • Naboulsi, W. et al., J Proteome.Res (2015)
    • Naderi, A., Exp.Cell Res 331 (2015): 239-250
    • Nakao, K. et al., J Gastroenterol. 49 (2014): 589-593
    • Navara, C. S., Curr.Pharm.Des 10 (2004): 1739-1744
    • Naz, S. et al., Carcinogenesis 35 (2014): 14-23
    • Neumann, M. et al., Blood 121 (2013): 4749-4752
    • Ng, S. K. et al., Clin Experiment.Ophthalmol. 43 (2015): 367-376
    • Nikitakis, N. G. et al., Am.J Clin Pathol. 119 (2003): 574-586
    • Nishida, C. R. et al., Mol.Pharmacol. 78 (2010): 497-502
    • Nwosu, V. et al., Hum.Mol Genet. 10 (2001): 2313-2318
    • Nykopp, T. K. et al., BMC.Cancer 10 (2010): 512
    • O'Gorman, D. B. et al., Endocrinology 143 (2002): 4287-4294
    • Oh, H. R. et al., Cell Oncol (Dordr.) 37 (2014): 455-461
    • Ohigashi, Y. et al., Clin Cancer Res. 11 (2005): 2947-2953
    • Okayama, H. et al., Cancer Epidemiol.Biomarkers Prev. 23 (2014): 2884-2894
    • Okoh, V. O. et al., PLoS.One. 8 (2013): e54206
    • Olstad, O. K. et al., Anticancer Res 23 (2003): 2201-2216
    • Ordonez, N. G., Arch.Pathol.Lab Med. 129 (2005): 1407-1414
    • Orzol, P. et al., Histol.Histopathol. 30 (2015): 503-521
    • Padden, J. et al., Mol.Cell Proteomics. 13 (2014): 2661-2672
    • Pan, B. et al., Mol.Biol Rep. 40 (2013): 27-33
    • Pan, T. et al., Biochem.Biophys.Res Commun. 456 (2015): 452-458
    • Papagerakis, S. et al., Hum.Pathol. 34 (2003): 565-572
    • Park, Y. et al., Oncogene 34 (2015): 5037-5045
    • Parker, L. P. et al., Cancer Genomics Proteomics. 6 (2009): 189-194
    • Pathak, S. et al., Nutr.Cancer 66 (2014): 818-824
    • Peng, H. et al., Cell Oncol (Dordr.) 38 (2015): 165-172
    • Penney, K. L. et al., Cancer Epidemiol.Biomarkers Prev. 24 (2015): 255-260
    • Perez, I. et al., Int.J Med.Sci. 12 (2015): 458-467
    • Persson, F. et al., Cancer Lett. 260 (2008): 37-47
    • Pflueger, D. et al., Neoplasia. 15 (2013): 1231-1240
    • Pickering, C. R. et al., Clin Cancer Res 20 (2014): 6582-6592
    • Pillay, V. et al., S.Afr.Med.J 105 (2015): 656-658
    • Pils, D. et al., BMC.Cancer 13 (2013): 178
    • Pinheiro, J. et al., nlme: Linear and Nonlinear Mixed Effects Models (http://CRAN.R-project.org/packe=nlme) (2015)
    • Plebanski, M. et al., Eur.J Immunol 25 (1995): 1783-1787
    • Porta, C. et al., Virology 202 (1994): 949-955
    • Prasad, N. B. et al., Mod.Pathol. 27 (2014): 945-957
    • Puente, X. S. et al., Nature 526 (2015): 519-524
    • Qendro, V. et al., J Proteome.Res 13 (2014): 5031-5040
    • Qi, Y. et al., Proteomics. 5 (2005): 2960-2971
    • Qi, Y. et al., J Breast Cancer 18 (2015): 218-224
    • Qie, S. et al., J Cell Biochem. 115 (2014): 498-509
    • Qu, Y. M. et al., Zhonghua Yi.Xue.Za Zhi. 90 (2010): 1958-1962
    • Qu, Z. et al., Cancer Med. 3 (2014): 453-461
    • Quillien, V. et al., Anticancer Res. 17 (1997): 387-391
    • Rabinovitz, I. et al., Biochem.Cell Biol 74 (1996): 811-821
    • Rabinovitz, I. et al., Clin Exp.Metastasis 13 (1995): 481-491
    • Rad, E. et al., Mol.Cancer Res 13 (2015): 1149-1160
    • Rai, R. et al., Oral Oncol 40 (2004): 705-712
    • Raica, M. et al., Anticancer Res 28 (2008): 2997-3006
    • Ramirez-Exposito, M. J. et al., Maturitas 72 (2012): 79-83
    • Rammensee, H. G. et al., Immunogenetics 50 (1999): 213-219
    • Reeb, A. N. et al., J Clin Endocrinol.Metab 100 (2015): E232-E242
    • RefSeq, The NCBI handbook [Internet], Chapter 18, (2002), http://www.ncbi.nlm.nih.gov/books/NBK21091 /
    • Rehman, I. et al., PLoS.One. 7 (2012): e30885
    • Reis, S. T. et al., Clinics.(Sao Paulo) 68 (2013): 652-657
    • Remmelink, M. et al., Int.J Oncol 26 (2005): 247-258
    • Revill, K. et al., Gastroenterology 145 (2013): 1424-1435
    • Ricketts, C. J. et al., Clin Epigenetics. 5 (2013): 16
    • Rini, B. I. et al., Cancer 107 (2006): 67-74
    • Rock, K. L. et al., Science 249 (1990): 918-921
    • Rodenko, B. et al., Nat Protoc. 1 (2006): 1120-1132
    • Roemer, A. et al., J Urol. 172 (2004): 2162-2166
    • Romana, S. P. et al., Leukemia 20 (2006): 696-706
    • Rozenblum, E. et al., Hum.Genet. 110 (2002): 111-121
    • Ruminy, P. et al., Leukemia 25 (2011): 681-688
    • Safadi, R. A. et al., Oral Surg.Oral Med.Oral Pathol.Oral Radiol. 121 (2016): 402-411
    • Saiki, R. K. et al., Science 239 (1988): 487-491
    • Sanchez-Palencia, A. et al., Int.J Cancer 129 (2011): 355-364
    • Santarpia, L. et al., Oncologist. 18 (2013): 1063-1073
    • Sarma, S. N. et al., Environ.Toxicol.Pharmacol. 32 (2011): 285-295
    • Sathyanarayana, U. G. et al., Cancer Res 64 (2004): 1425-1430
    • Sato, T. et al., Oncogene 33 (2014): 2215-2224
    • Sato, Y. et al., J Gastroenterol.Hepatol. 28 (2013): 1422-1429
    • Savaskan, N. E. et al., Ann.Anat. 192 (2010): 309-313
    • Savaskan, N. E. et al., Curr.Neuropharmacol. 13 (2015): 258-265
    • Savoy, R. M. et al., Endocr.Relat Cancer 20 (2013): R341-R356
    • Schlieben, P. et al., Vet.J 194 (2012): 210-214
    • Schmitt-Graeff, A. et al., Histopathology 51 (2007): 87-97
    • Schuld, N. J. et al., Cell Cycle 13 (2014): 941-952
    • Schumann, H. et al., Br.J Dermatol. 167 (2012): 929-936
    • Scrideli, C. A. et al., J Neurooncol. 88 (2008): 281-291
    • Seda, V. et al., Eur.J Haematol. 94 (2015): 193-205
    • Seeger, F. H. et al., Immunogenetics 49 (1999): 571-576
    • Seitz, S. et al., Eur.J Cancer 36 (2000): 1507-1513
    • Semenza, G. L., Cold Spring Harb.Symp.Quant.Biol 76 (2011): 347-353
    • Seong, J. et al., Mol.Biol.Rep. 39 (2012): 3597-3601
    • Sethi, M. K. et al., J Proteomics. 126 (2015): 54-67
    • Sherman, F. et al., Laboratory Course Manual for Methods in Yeast Genetics (1986)
    • Shi, Z. et al., Int.J Gynecol.Cancer 22 (2012): 1125-1129
    • Shi, Z. G. et al., Clin Transl.Oncol 17 (2015): 65-73
    • Shibano, T. et al., PLoS.One. 10 (2015): e0127271
    • Shin, S. H. et al., Lab Invest 94 (2014): 1396-1405
    • Shruthi, D. K. et al., J Oral Maxillofac.Pathol. 18 (2014): 365-371
    • Silva, J. M. et al., Cell 137 (2009): 1047-1061
    • Silva, L. P. et al., Anal.Chem. 85 (2013): 9536-9542
    • Singh, V. et al., OMICS. 19 (2015): 688-699
    • Singh-Jasuja, H. et al., Cancer Immunol.Immunother. 53 (2004): 187-195
    • Slaga, T. J. et al., J Investig.Dermatol.Symp.Proc. 1 (1996): 151-156
    • Small, E. J. et al., J Clin Oncol. 24 (2006): 3089-3094
    • Sobolik-Delmaire, T. et al., Cell Commun.Adhes. 14 (2007): 99-109
    • Spurr, I. B. et al., Chembiochem. 13 (2012): 1628-1634
    • Stahl, M. et al., Ann.Oncol. 24 Suppl 6 (2013): vi51-vi56
    • Stull, R. A. et al., BMC.Genomics 6 (2005): 55
    • Sturm, M. et al., BMC.Bioinformatics. 9 (2008): 163
    • Sugimoto, K. J. et al., Int.J Clin Exp.Pathol. 7 (2014): 8980-8987
    • Suh, J. H. et al., J Korean Med.Sci. 28 (2013): 593-601
    • Sun, M. et al., Biochem.Biophys.Res Commun. 340 (2006): 209-214
    • Sun, Y. et al., Biochem.Biophys.Res Commun. 450 (2014): 1-6
    • Suzuki, S. et al., Pathol.Res Pract. 210 (2014): 130-134
    • Swain, N. et al., Tumour.Biol 35 (2014): 8407-8413
    • Szeliga, M. et al., Tumour.Biol 35 (2014): 1855-1862
    • Takabe, P. et al., Exp.Cell Res 337 (2015): 1-15
    • Takahashi, H. et al., Urology 79 (2012): 240-248
    • Takeda, H. et al., Nat Genet. 47 (2015): 142-150
    • Tamada, M. et al., Clin Cancer Res 18 (2012): 5554-5561
    • Tan, B. S. et al., Mol.Cancer Ther. 10 (2011): 1982-1992
    • Tanaka, F. et al., Int.J Oncol 10 (1997): 1113-1117
    • Tang, H. et al., Anticancer Drugs 18 (2007): 633-639
    • Tang, H. et al., Clin Cancer Res 19 (2013): 1577-1586
    • Tang, J. Q. et al., Beijing Da.Xue.Xue.Bao. 41 (2009): 531-536
    • Tang, J. Q. et al., Chin Med.J (Engl.) 123 (2010): 3559-3565
    • Tanis, T. et al., Arch.Oral Biol 59 (2014): 1155-1163
    • Tech, K. et al., Cancer Lett. 356 (2015): 268-272
    • Teng, B. P. et al., Anticancer Agents Med.Chem 11 (2011): 620-628
    • Terada, T., Int.J Clin Exp.Pathol. 5 (2012): 596-600
    • Teufel, R. et al., Cell Mol Life Sci. 62 (2005): 1755-1762
    • Tew, G. W. et al., J Biol Chem 283 (2008): 963-976
    • Thomas, A. et al., Cancer Med. 2 (2013): 836-848
    • Tian, S. Y. et al., Int.J Clin Exp.Pathol. 7 (2014): 3752-3762
    • Tofuku, K. et al., Int.J Oncol 29 (2006): 175-183
    • Toh, U. et al., Int.J Clin Oncol 7 (2002): 372-375
    • Toh, U. et al., Clin Cancer Res. 6 (2000): 4663-4673
    • Toomey, P. G. et al., Cancer Control 20 (2013): 32-42
    • Tota, G. et al., BMC.Cancer 14 (2014): 963
    • Tran, E. et al., Science 344 (2014): 641-645
    • Truong, T. et al., Endocr.Relat Cancer 21 (2014): 629-638
    • Tsujimoto, H. et al., Mol.Carcinog 26 (1999): 298-304
    • Tuupanen, S. et al., Br.J Cancer 111 (2014): 1657-1662
    • Tuval-Kochen, L. et al., PLoS.One. 8 (2013): e77260
    • Twa, D. D. et al., J Pathol. 236 (2015): 136-141
    • Twarock, S. et al., Mol.Cancer 10 (2011): 30
    • Tzellos, T. G. et al., J Eur.Acad.Dermatol.Venereol. 25 (2011): 679-687
    • Urosevic, J. et al., Nat Cell Biol 16 (2014): 685-694
    • Vachani, A. et al., Clin Cancer Res. 13 (2007): 2905-2915
    • Valladares-Ayerbes, M. et al., Cancer Epidemiol.Biomarkers Prev. 19 (2010): 1432-1440
    • Valletta, D. et al., Carcinogenesis 35 (2014): 1407-1415
    • van, Geldermalsen M. et al., Oncogene (2015)
    • Varga, A. E. et al., Oncogene 24 (2005): 5043-5052
    • Varona, A. et al., Am.J Physiol Renal Physiol 292 (2007): F780-F788
    • Vasca, V. et al., Oncol Lett. 8 (2014): 2501-2504
    • Venneti, S. et al., Brain Pathol. 23 (2013): 217-221
    • Virtakoivu, R. et al., Cancer Res 75 (2015): 2349-2362
    • Volkmer, J. P. et al., Proc.Natl.Acad.Sci.U.S.A 109 (2012): 2078-2083
    • Volpi, A. et al., G.Chir 32 (2011): 59-63
    • Vui-Kee, K. et al., Kaohsiung.J Med.Sci. 28 (2012): 243-250
    • Walter, S. et al., J Immunol 171 (2003): 4974-4978
    • Walter, S. et al., Nat Med. 18 (2012): 1254-1261
    • Wang, D. et al., Biochem.Biophys.Res Commun. 458 (2015a): 313-320
    • Wang, H. et al., Front Oncol 4 (2014): 377
    • Wang, H. et al., Cancer Cell 18 (2010): 52-62
    • Wang, J. et al., Oncol Rep. 33 (2015b): 1326-1334
    • Wang, T. et al., Tumour.Biol (2015c)
    • Wang, W. M. et al., J Biol Chem 278 (2003): 19549-19557
    • Wang, X. et al., Eur.J Pharmacol. 768 (2015d): 116-122
    • Wang, X. M. et al., PLoS.One. 8 (2013a): e55714
    • Wang, X. Y. et al., Int J Hyperthermia 29 (2013): 364-375
    • Wang, Y. et al., Neoplasma 62 (2015e): 966-973
    • Wang, Z. et al., Oncotarget. 4 (2013b): 2476-2486
    • Wang, Z. et al., Melanoma Res 14 (2004): 107-114
    • Warner, S. L. et al., Future.Med.Chem 6 (2014): 1167-1178
    • Watanabe, Y. et al., Gastroenterology 136 (2009): 2149-2158
    • Wegdam, W. et al., PLoS.One. 9 (2014): e108046
    • Wehner, M. et al., FEBS J 277 (2010): 1597-1605
    • Weiss, I. et al., Int.J Mol.Sci. 13 (2012): 12925-12938
    • Weissbach, S. et al., Br.J Haematol. 169 (2015): 57-70
    • Wiedl, T. et al., J Proteomics. 74 (2011): 1884-1894
    • Willcox, B. E. et al., Protein Sci. 8 (1999): 2418-2423
    • Willoughby, V. et al., Appl.Immunohistochem.Mol.Morphol. 16 (2008): 344-348
    • Wittke, I. et al., Cancer Lett. 162 (2001): 237-243
    • Wojtalewicz, N. et al., PLoS.One. 9 (2014): e90461
    • Wong, N. et al., Cancer Lett. 356 (2015): 184-191
    • Woo, T. et al., PLoS.One. 10 (2015): e0142642
    • World Cancer Report, (2014)
    • Wu, G. et al., Onco.Targets.Ther. 8 (2015): 2067-2074
    • Wu, S. et al., Acta Biochim.Biophys.Sin.(Shanghai) 45 (2013): 27-35
    • Wu, X. et al., Cancer Res 70 (2010): 2718-2727
    • Xiang, Y. et al., J Clin Invest 125 (2015): 2293-2306
    • Xu, J. et al., Genet.Mol.Res 13 (2014): 5732-5744
    • Xu, X. et al., Oncotarget. 6 (2015): 26161-26176
    • Xue, L. Y. et al., Zhonghua Zhong.Liu Za Zhi. 32 (2010): 838-844
    • Yager, M. L. et al., Br.J Cancer 89 (2003): 860-863
    • Yamaguchi, T. et al., Dis.Colon Rectum 49 (2006): 399-406
    • Yamamoto, M. et al., PLoS.One. 6 (2011): e17149
    • Yamamoto, N. et al., Int.J Oncol 42 (2013): 1523-1532
    • Yang, C. et al., Tumour.Biol (2015a)
    • Yang, C. et al., Exp.Cell Res 331 (2015b): 377-386
    • Yang, H. Y. et al., J Proteomics. 75 (2012): 3639-3653
    • Yang, J. Y. et al., BMC.Cancer 10 (2010): 388
    • Yang, S. et al., J Cancer Res Clin Oncol 141 (2015c): 1265-1275
    • Yang, W. et al., Cancer Lett. 339 (2013): 153-158
    • Yang, W. et al., Nature 499 (2013a): 491-495
    • Yang, W. et al., Int.J Oncol 42 (2013b): 690-698
    • Yao, M. et al., Cancer Med. 3 (2014): 845-854
    • Yao, R. et al., Histol.Histopathol. 22 (2007): 1025-1032
    • Yu, D. et al., Oncotarget. 6 (2015a): 7619-7631
    • Yu, X. et al., Cancer Res 73 (2013): 2093-2103
    • Yu, Y. et al., Cancer Cell 28 (2015b): 82-96
    • Yuan, B. et al., Immunobiology 217 (2012): 738-742
    • Zang, W. et al., Mol.Cancer 14 (2015): 37
    • Zanini, S. et al., Cell Signal. 27 (2015): 899-907
    • Zare, M. et al., Mol.Carcinog 51 (2012): 796-806
    • Zaremba, S. et al., Cancer Res. 57 (1997): 4570-4577
    • Zha, C. et al., PLoS.One. 10 (2015): e0122322
    • Zhang, D. et al., J Cell Mol.Med. 16 (2012): 1047-1059
    • Zhang, H. Y. et al., Mol.Biol Rep. 41 (2014): 5519-5524
    • Zhang, Q. et al., J Cancer Res Clin Oncol 141 (2015a): 691-703
    • Zhang, S. et al., Cancer Res 64 (2004): 2977-2983
    • Zhang, S. et al., J Mol.Histol. 45 (2014): 283-292
    • Zhang, S. N. et al., Zhonghua Yi.Xue.Za Zhi. 85 (2005): 1348-1351
    • Zhang, T. et al., Mol.Cancer 9 (2010): 72
    • Zhang, X. et al., Int.J Cancer 137 (2015b): 2803-2814
    • Zhang, X. et al., Tumour.Biol 36 (2015c): 5979-5985
    • Zhang, X. et al., PLoS.One. 8 (2013): e72458
    • Zhang, Y. et al., Cancer Metastasis Rev 34 (2015d): 249-264
    • Zhang, Z. Z. et al., Mol.Cancer Ther. 14 (2015e): 1162-1170
    • Zhao, D. et al., J Neurooncol. 118 (2014a): 39-47
    • Zhao, G. et al., Biochem.Biophys.Res Commun. 408 (2011): 154-159
    • Zhao, H. et al., Gene 548 (2014b): 234-243
    • Zhao, L. J. et al., Chin Med.J (Engl.) 126 (2013): 4260-4264
    • Zheng, Q. et al., Tumour.Biol 35 (2014): 6255-6264
    • Zheng, R. et al., Int.Immunopharmacol. 29 (2015): 919-925
    • Zhi, H. et al., J Pathol. 217 (2009): 389-397
    • Zhou, Y. F. et al., World J Gastroenterol. 20 (2014): 13172-13177
    • Zhu, H. et al., Cancer Lett. 245 (2007a): 303-314
    • Zhu, L. et al., J Dermatol.Sci. 72 (2013a): 311-319
    • Zhu, S. et al., J Biol Chem 282 (2007b): 14328-14336
    • Zhu, Y. et al., Prostate 73 (2013b): 1614-1622
    • Zhu, Y. P. et al., Oncotarget. 6 (2015): 14488-14496
    SEQUENCE LISTING
    • <110> immatics biotechnologies GmbH
    • <120> Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
    • <130> I32873WO
    • <150> US62/188,870
      <151> 2015-07-06
    • <150> GB1511792.2
      <151> 2015-07-06
    • <160> 103
    • <170> PatentIn version 3.5
    • <210> 1
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 1
      Figure imgb0002
    • <210> 2
      <211> 13
      <212> PRT
      <213> Homo sapiens
    • <400> 2
      Figure imgb0003
    • <210> 3
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 3
      Figure imgb0004
    • <210> 4
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 4
      Figure imgb0005
    • <210> 5
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 5
      Figure imgb0006
    • <210> 6
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 6
      Figure imgb0007
    • <210> 7
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 7
      Figure imgb0008
    • <210> 8
      <211> 13
      <212> PRT
      <213> Homo sapiens
    • <400> 8
      Figure imgb0009
    • <210> 9
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 9
      Figure imgb0010
    • <210> 10
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 10
      Figure imgb0011
      Figure imgb0012
    • <210> 11
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 11
      Figure imgb0013
    • <210> 12
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 12
      Figure imgb0014
    • <210> 13
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 13
      Figure imgb0015
    • <210> 14
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 14
      Figure imgb0016
    • <210> 15
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 15
      Figure imgb0017
    • <210> 16
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 16
      Figure imgb0018
    • <210> 17
      <211> 8
      <212> PRT
      <213> Homo sapiens
    • <400> 17
      Figure imgb0019
    • <210> 18
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 18
      Figure imgb0020
    • <210> 19
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 19
      Figure imgb0021
    • <210> 20
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 20
      Figure imgb0022
    • <210> 21
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 21
      Figure imgb0023
    • <210> 22
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 22
      Figure imgb0024
    • <210> 23
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 23
      Figure imgb0025
    • <210> 24
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 24
      Figure imgb0026
    • <210> 25
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 25
      Figure imgb0027
    • <210> 26
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 26
      Figure imgb0028
    • <210> 27
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 27
      Figure imgb0029
    • <210> 28
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 28
      Figure imgb0030
    • <210> 29
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 29
      Figure imgb0031
    • <210> 30
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 30
      Figure imgb0032
    • <210> 31
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 31
      Figure imgb0033
    • <210> 32
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 32
      Figure imgb0034
    • <210> 33
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 33
      Figure imgb0035
    • <210> 34
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 34
      Figure imgb0036
    • <210> 35
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 35
      Figure imgb0037
    • <210> 36
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 36
      Figure imgb0038
    • <210> 37
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 37
      Figure imgb0039
    • <210> 38
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 38
      Figure imgb0040
    • <210> 39
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 39
      Figure imgb0041
    • <210> 40
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 40
      Figure imgb0042
    • <210> 41
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 41
      Figure imgb0043
    • <210> 42
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 42
      Figure imgb0044
    • <210> 43
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 43
      Figure imgb0045
    • <210> 44
      <211> 14
      <212> PRT
      <213> Homo sapiens
    • <400> 44
      Figure imgb0046
    • <210> 45
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 45
      Figure imgb0047
    • <210> 46
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 46
      Figure imgb0048
    • <210> 47
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 47
      Figure imgb0049
    • <210> 48
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 48
      Figure imgb0050
    • <210> 49
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 49
      Figure imgb0051
    • <210> 50
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 50
      Figure imgb0052
    • <210> 51
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 51
      Figure imgb0053
    • <210> 52
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 52
      Figure imgb0054
    • <210> 53
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 53
      Figure imgb0055
    • <210> 54
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 54
      Figure imgb0056
    • <210> 55
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 55
      Figure imgb0057
    • <210> 56
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 56
      Figure imgb0058
    • <210> 57
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 57
      Figure imgb0059
    • <210> 58
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 58
      Figure imgb0060
    • <210> 59
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 59
      Figure imgb0061
    • <210> 60
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 60
      Figure imgb0062
    • <210> 61
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 61
      Figure imgb0063
    • <210> 62
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 62
      Figure imgb0064
    • <210> 63
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 63
      Figure imgb0065
    • <210> 64
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 64
      Figure imgb0066
    • <210> 65
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 65
      Figure imgb0067
    • <210> 66
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 66
      Figure imgb0068
    • <210> 67
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 67
      Figure imgb0069
    • <210> 68
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 68
      Figure imgb0070
    • <210> 69
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 69
      Figure imgb0071
    • <210> 70
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 70
      Figure imgb0072
    • <210> 71
      <211> 12
      <212> PRT
      <213> Homo sapiens
    • <400> 71
      Figure imgb0073
    • <210> 72
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 72
      Figure imgb0074
    • <210> 73
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 73
      Lys Ile Thr Glu Ala Ile Gln Tyr Val
      Figure imgb0075
    • <210> 74
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 74
      Figure imgb0076
    • <210> 75
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 75
      Figure imgb0077
    • <210> 76
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 76
      Figure imgb0078
    • <210> 77
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 77
      Figure imgb0079
    • <210> 78
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 78
      Figure imgb0080
    • <210> 79
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 79
      Figure imgb0081
    • <210> 80
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 80
      Figure imgb0082
    • <210> 81
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 81
      Figure imgb0083
    • <210> 82
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 82
      Figure imgb0084
    • <210> 83
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 83
      Figure imgb0085
    • <210> 84
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 84
      Figure imgb0086
    • <210> 85
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 85
      Figure imgb0087
    • <210> 86
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 86
      Figure imgb0088
    • <210> 87
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 87
      Figure imgb0089
    • <210> 88
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 88
      Figure imgb0090
    • <210> 89
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 89
      Figure imgb0091
    • <210> 90
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 90
      Figure imgb0092
    • <210> 91
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 91
      Figure imgb0093
    • <210> 92
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 92
      Figure imgb0094
    • <210> 93
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 93
      Figure imgb0095
    • <210> 94
      <211> 11
      <212> PRT
      <213> Homo sapiens
    • <400> 94
      Figure imgb0096
    • <210> 95
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 95
      Figure imgb0097
    • <210> 96
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 96
      Figure imgb0098
    • <210> 97
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 97
      Figure imgb0099
    • <210> 98
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 98
      Figure imgb0100
    • <210> 99
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 99
      Figure imgb0101
    • <210> 100
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 100
      Figure imgb0102
    • <210> 101
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 101
      Figure imgb0103
    • <210> 102
      <211> 10
      <212> PRT
      <213> Homo sapiens
    • <400> 102
      Figure imgb0104
    • <210> 103
      <211> 9
      <212> PRT
      <213> Homo sapiens
    • <400> 103
      Figure imgb0105

Claims (13)

  1. A peptide consisting of the amino acid sequence according to SEQ ID No. 9; or a pharmaceutical acceptable salt thereof for use in medicine.
  2. The peptide for use according to claim 1, wherein said peptide includes non-peptide bonds.
  3. The peptide for use according to claim 1 or 2, wherein said peptide is part of a fusion protein comprising the N-terminal 80 amino acids of the HLA-DR antigen-associated invariant chain (Ii).
  4. A T-cell receptor, preferably a recombinant, soluble or membrane-bound T-cell receptor that is specifically reactive with an HLA ligand when bound to an HLA molecule, wherein said ligand consists of the amino acid sequence of SEQ ID No. 9.
  5. An antibody, in particular a soluble or membrane-bound antibody that specifically recognizes the peptide for use according to claim 1, preferably the peptide for use according to claim 1 when bound to an MHC molecule.
  6. A nucleic acid, encoding for a peptide for use according to any one of claims 1 or 3, for a TCR according to claim 4, or an antibody according to claim 5, optionally linked to a heterologous promoter sequence, or an expression vector expressing said nucleic acid.
  7. A recombinant host cell comprising the peptide for use according to claim 1 or 3, or the nucleic acid or the expression vector according to claim 6, wherein said host cell preferably is an antigen presenting cell such as a dendritic cell, or wherein said host cell preferably is a T cell or NK cell.
  8. A method for producing the peptide for use according to any one of claims 1 or 3, or for producing the T cell receptor according to claim 4, or an antibody according to claim 5, the method comprising culturing the host cell according to claim 7 that presents the peptide for use according to claim 1, or expresses the nucleic acid or expression vector according to claim 6, and isolating the peptide or the TCR or the antibody from the host cell or its culture medium.
  9. An in vitro method for producing activated T lymphocytes, the method comprising contacting in vitro T cells with antigen loaded human class I MHC molecules expressed on the surface of a suitable antigen-presenting cell or an artificial construct mimicking an antigen-presenting cell for a period of time sufficient to activate said T cells in an antigen specific manner, wherein said antigen is a peptide for use according to claim 1.
  10. An activated T lymphocyte, produced by the method according to claim 9 that selectively recognizes a cell which presents a polypeptide comprising an amino acid sequence given in claim 1.
  11. A pharmaceutical composition comprising at least one active ingredient selected from the group consisting of the peptide for use according to any one of claims 1 or 3, the nucleic acid or the expression vector according to claim 6, the cell according to claim 7, the activated T lymphocyte according to claim 10 or the antibody according to claim 5 or T-cell receptor according to claim 4, and a pharmaceutically acceptable carrier, and optionally additional pharmaceutically acceptable excipients and/or stabilizers for use in medicine.
  12. The peptide for use according to any one of claims 1 or 3, the nucleic acid or the expression vector according to claim 6, the cell according to claim 7, the activated T lymphocyte according to claim 10 or the antibody according to claim 5, the T-cell receptor according to claim 4 or the pharmaceutical composition for use according to claim 11 for use in medicine, preferably for use in the treatment of cancer.
  13. The peptide for use according to any one of claims 1 or 3, the nucleic acid or the expression vector according to claim 6, the cell according to claim 7, the activated T lymphocyte according to claim 10 or the antibody according to claim 5, the T-cell receptor according to claim 4 or the pharmaceutical composition for use according to claim 11 for use according to claim 12, wherein said cancer is selected from the group of ovarian cancer, non-small cell lung cancer, small cell lung cancer, kidney cancer, brain cancer, colon or rectum cancer, stomach cancer, liver cancer, pancreatic cancer, prostate cancer, leukemia, breast cancer, Merkel cell carcinoma, melanoma, esophageal cancer, urinary bladder cancer, uterine cancer, gallbladder cancer, bile duct cancer and other tumors that show an overexpression of a protein from which a peptide according to SEQ ID No. 9 is derived from.
EP16736073.4A 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers Active EP3319985B1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP20175260.7A EP3736283A1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
SI201631018T SI3319985T1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
EP23216397.2A EP4321172A2 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
RS20201531A RS61203B1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
PL16736073T PL3319985T3 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
HRP20210076TT HRP20210076T1 (en) 2015-07-06 2021-01-14 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1511792.2A GB201511792D0 (en) 2015-07-06 2015-07-06 Novel peptides and combination of peptides for use in immunotherapy against esopageal cancer and other cancers
PCT/EP2016/065812 WO2017005733A2 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Related Child Applications (3)

Application Number Title Priority Date Filing Date
EP20175260.7A Division EP3736283A1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
EP20175260.7A Division-Into EP3736283A1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
EP23216397.2A Division EP4321172A2 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Publications (2)

Publication Number Publication Date
EP3319985A2 EP3319985A2 (en) 2018-05-16
EP3319985B1 true EP3319985B1 (en) 2020-10-28

Family

ID=54013532

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16736073.4A Active EP3319985B1 (en) 2015-07-06 2016-07-05 Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Country Status (7)

Country Link
US (10) US10011645B2 (en)
EP (1) EP3319985B1 (en)
CY (1) CY1123902T1 (en)
GB (1) GB201511792D0 (en)
MA (2) MA41522A1 (en)
TW (1) TWI765857B (en)
WO (1) WO2017005733A2 (en)

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180107102A (en) 2015-12-16 2018-10-01 그릿스톤 온콜로지, 인코포레이티드 Identification of new antigens, manufacture, and uses
GB201603568D0 (en) * 2016-03-01 2016-04-13 Immatics Biotechnologies Gmbh Efficient treatment options including peptides and combination of peptide and cell based medicaments for use in immunotherapy against urinary bladder cancer
JP7075125B2 (en) 2016-05-25 2022-05-25 イマティクス バイオテクノロジーズ ゲーエムベーハー A novel peptide, peptide combination as a target and for use in immunotherapy for gallbladder cancer and cholangiocarcinoma and other cancers
GB201609193D0 (en) 2016-05-25 2016-07-06 Immatics Biotechnologies Gmbh Novel peptides, combination of peptides as targets for use in immunotherapy against gallbladder cancer and cholangiocarcinoma and other cancers
TWI796299B (en) * 2016-08-26 2023-03-21 德商英麥提克生物技術股份有限公司 Novel peptides and scaffolds for use in immunotherapy against head and neck squamous cell carcinoma and other cancers
EP4317432A3 (en) 2016-12-08 2024-04-17 Immatics Biotechnologies GmbH T cell receptors with improved pairing
DE102016123893A1 (en) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T cell receptors with improved binding
DE102017115966A1 (en) 2017-07-14 2019-01-17 Immatics Biotechnologies Gmbh Polypeptide molecule with improved dual specificity
SG11202000027WA (en) 2017-07-14 2020-02-27 Immatics Biotechnologies Gmbh Improved dual specificity polypeptide molecule
CN111465989A (en) 2017-10-10 2020-07-28 磨石肿瘤生物技术公司 Identification of neoantigens using hot spots
EP3714275A4 (en) 2017-11-22 2021-10-27 Gritstone bio, Inc. Reducing junction epitope presentation for neoantigens
DE102017127984B4 (en) 2017-11-27 2019-12-05 Immatics US, Inc. Method for the propagation and activation of γδ T cells
KR20200118450A (en) * 2018-02-05 2020-10-15 아이벡스 바이오사이언시스, 엘엘씨 Use of PCBP1 to treat hyperproliferative diseases
CN111684062A (en) 2018-02-09 2020-09-18 伊玛提克斯美国公司 Method for producing T cell
KR102073144B1 (en) * 2018-12-13 2020-02-04 주식회사 엘베이스 Pharmaceutical Composition for Preventing or Treating Cancer Comprising Oligopeptide
US20200297768A1 (en) 2019-03-19 2020-09-24 Immatics US, Inc. Cd28 t cell cultures, compositions, and methods of using thereof
EA202193139A1 (en) 2019-05-27 2022-03-01 Имматикс Юс, Инк. VIRAL VECTORS AND THEIR APPLICATIONS IN ADOPTIONAL CELL THERAPY
KR20220018006A (en) 2019-06-06 2022-02-14 이매틱스 바이오테크놀로지스 게엠베하 Classification by counter selection using sequence-like peptides
US20210032370A1 (en) 2019-08-02 2021-02-04 Immatics Biotechnologies Gmbh Recruiting agent further binding an mhc molecule
IL295891A (en) 2020-02-24 2022-10-01 Immatics Us Inc Methods for expanding t cells for the treatment of cancer and related malignancies
DE102020111571A1 (en) 2020-03-11 2021-09-16 Immatics US, Inc. WPRE MUTANT CONSTRUCTS, COMPOSITIONS, AND RELATED PROCEDURES
DE102020106710A1 (en) 2020-03-11 2021-09-16 Immatics US, Inc. WPRE MUTANT CONSTRUCTS, COMPOSITIONS, AND RELATED PROCEDURES
WO2022040631A1 (en) 2020-08-21 2022-02-24 Immatics US, Inc. Methods for isolating cd8+ selected t cells
CN114317730B (en) * 2020-09-29 2024-01-09 香港中文大学深圳研究院 Application of TTPAL as gastric cancer monitoring or prognosis judgment marker
JP2024502034A (en) 2020-12-31 2024-01-17 イマティクス ユーエス,アイエヌシー. CD8 polypeptides, compositions, and methods of using them
BR112023022975A2 (en) 2021-05-05 2024-01-23 Immatics Biotechnologies Gmbh IMPROVED BMA031 ANTIGEN BINDING POLYPEPTIDES
WO2023025851A1 (en) 2021-08-24 2023-03-02 Immatics US, Inc. Selection of immune cells using peptide mhc complexes generated by conditional ligand exchange
TW202332765A (en) 2021-09-20 2023-08-16 美商英麥提克斯股份有限公司 Monocyte depletion of t cells populations for t-cell therapy
WO2023081925A1 (en) 2021-11-08 2023-05-11 Immatics Biotechnologies Gmbh Adoptive cell therapy combination treatment and compositions thereof
US20240066127A1 (en) 2022-04-28 2024-02-29 Immatics US, Inc. Il-12 polypeptides, il-15 polypeptides, il-18 polypeptides, cd8 polypeptides, compositions, and methods of using thereof
WO2023212697A1 (en) 2022-04-28 2023-11-02 Immatics US, Inc. Membrane-bound il-15, cd8 polypeptides, cells, compositions, and methods of using thereof
US20230348561A1 (en) 2022-04-28 2023-11-02 Immatics US, Inc. Dominant negative tgfbeta receptor polypeptides, cd8 polypeptides, cells, compositions, and methods of using thereof
WO2023215825A1 (en) 2022-05-05 2023-11-09 Immatics US, Inc. Methods for improving t cell efficacy

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7103198A (en) * 1997-04-21 1998-11-13 Schering Corporation Mammalian cytokines; related reagents and methods
JP2001512013A (en) * 1997-08-01 2001-08-21 ジェンセット 5'EST of secreted protein expressed in prostate
AU2001219052A1 (en) * 2000-11-10 2002-05-21 Alexei Alexandrovich Gorchakov Portable computer apparatus
US7378495B2 (en) * 2002-10-21 2008-05-27 Pevion Biotech, Ltd. PTH-rP related peptide cancer therapeutics
US20040220094A1 (en) * 2003-05-01 2004-11-04 Skinner Keith K. Inverse agonist and agonist peptides that stimulate/inhibit hair growth
DE602005016112D1 (en) 2005-09-05 2009-10-01 Immatics Biotechnologies Gmbh Tumor-associated peptides that bind HLA class I or II molecules and anti-tumor vaccines
US20100129928A1 (en) 2006-07-27 2010-05-27 Roberto Polakewicz Tyrosine Phosphorylation Sites
GB201006360D0 (en) 2010-04-16 2010-06-02 Immatics Biotechnologies Gmbh Method for differentially quantifying naturally processed HLA-restricted peptides for cancer, autoimmune and infectious diseases immunotherapy development
GB201009222D0 (en) 2010-06-02 2010-07-21 Immatics Biotechnologies Gmbh Improved cancer therapy based on tumour associated antigens derived from cyclin D1

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
TW201713684A (en) 2017-04-16
EP3319985A2 (en) 2018-05-16
US20190177397A1 (en) 2019-06-13
US10011645B2 (en) 2018-07-03
US20190002525A1 (en) 2019-01-03
US10202437B2 (en) 2019-02-12
WO2017005733A3 (en) 2017-02-16
US20180282393A1 (en) 2018-10-04
US20180282394A1 (en) 2018-10-04
US10294288B2 (en) 2019-05-21
US10364282B2 (en) 2019-07-30
MA41522A1 (en) 2018-05-31
US10221229B2 (en) 2019-03-05
US10189886B2 (en) 2019-01-29
GB201511792D0 (en) 2015-08-19
WO2017005733A2 (en) 2017-01-12
US10273282B2 (en) 2019-04-30
CY1123902T1 (en) 2022-03-24
US20190010209A1 (en) 2019-01-10
US10239930B2 (en) 2019-03-26
MA42381B1 (en) 2020-12-31
US10214577B2 (en) 2019-02-26
US10167331B1 (en) 2019-01-01
US20180251516A1 (en) 2018-09-06
TWI765857B (en) 2022-06-01
US20170029486A1 (en) 2017-02-02
MA42381A (en) 2018-05-16
US20190010208A1 (en) 2019-01-10
US20190016779A1 (en) 2019-01-17
US20190002526A1 (en) 2019-01-03

Similar Documents

Publication Publication Date Title
EP3319985B1 (en) Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
AU2021200800B2 (en) Novel peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers
US11999774B2 (en) Peptides and combination of peptides for use in immunotherapy against esophageal cancer and other cancers

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180131

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: FRITSCHE, JENS

Inventor name: WEINSCHENK, TONI

Inventor name: SCHOOR, OLIVER

Inventor name: SONG, COLETTE

Inventor name: SINGH, HARPREET

Inventor name: MAHR, ANDREA

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1255542

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20191031

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20200506

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1328134

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201115

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602016046695

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: MA

Ref legal event code: VAGR

Ref document number: 42381

Country of ref document: MA

Kind code of ref document: B1

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20201222

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3319985

Country of ref document: PT

Date of ref document: 20210106

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20201229

REG Reference to a national code

Ref country code: FI

Ref legal event code: FGE

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20210076T

Country of ref document: HR

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E020072

Country of ref document: EE

Effective date: 20201123

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: MD

Ref legal event code: VAGR

Ref document number: 3319985

Country of ref document: MD

Date of ref document: 20210131

Kind code of ref document: T2

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 36000

Country of ref document: SK

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20210400033

Country of ref document: GR

Effective date: 20210215

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20201028

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20210076

Country of ref document: HR

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E052628

Country of ref document: HU

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2841507

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20210708

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602016046695

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210076

Country of ref document: HR

Payment date: 20210630

Year of fee payment: 6

VSFP Annual fee paid to validation state [announced via postgrant information from national office to epo]

Ref country code: MD

Payment date: 20210618

Year of fee payment: 6

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20210729

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

VSFP Annual fee paid to validation state [announced via postgrant information from national office to epo]

Ref country code: MD

Payment date: 20220616

Year of fee payment: 7

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210076

Country of ref document: HR

Payment date: 20220627

Year of fee payment: 7

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20210076

Country of ref document: HR

Payment date: 20230628

Year of fee payment: 8

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230526

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: RO

Payment date: 20230626

Year of fee payment: 8

Ref country code: PT

Payment date: 20230626

Year of fee payment: 8

Ref country code: LT

Payment date: 20230626

Year of fee payment: 8

Ref country code: CZ

Payment date: 20230623

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230626

Year of fee payment: 8

Ref country code: NL

Payment date: 20230720

Year of fee payment: 8

Ref country code: LU

Payment date: 20230719

Year of fee payment: 8

Ref country code: HR

Payment date: 20230628

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NO

Payment date: 20230720

Year of fee payment: 8

Ref country code: IT

Payment date: 20230731

Year of fee payment: 8

Ref country code: IE

Payment date: 20230726

Year of fee payment: 8

Ref country code: GB

Payment date: 20230724

Year of fee payment: 8

Ref country code: FI

Payment date: 20230719

Year of fee payment: 8

Ref country code: ES

Payment date: 20230821

Year of fee payment: 8

Ref country code: EE

Payment date: 20230718

Year of fee payment: 8

Ref country code: CY

Payment date: 20230623

Year of fee payment: 8

Ref country code: CH

Payment date: 20230801

Year of fee payment: 8

Ref country code: BG

Payment date: 20230719

Year of fee payment: 8

Ref country code: AT

Payment date: 20230718

Year of fee payment: 8

VSFP Annual fee paid to validation state [announced via postgrant information from national office to epo]

Ref country code: MD

Payment date: 20230619

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20230703

Year of fee payment: 8

Ref country code: SI

Payment date: 20230626

Year of fee payment: 8

Ref country code: SE

Payment date: 20230724

Year of fee payment: 8

Ref country code: RS

Payment date: 20230703

Year of fee payment: 8

Ref country code: PL

Payment date: 20230705

Year of fee payment: 8

Ref country code: IS

Payment date: 20230727

Year of fee payment: 8

Ref country code: HU

Payment date: 20230629

Year of fee payment: 8

Ref country code: GR

Payment date: 20230718

Year of fee payment: 8

Ref country code: FR

Payment date: 20230724

Year of fee payment: 8

Ref country code: DK

Payment date: 20230724

Year of fee payment: 8

Ref country code: DE

Payment date: 20230720

Year of fee payment: 8

Ref country code: BE

Payment date: 20230719

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20230721

Year of fee payment: 8

Ref country code: LV

Payment date: 20230719

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20230623

Year of fee payment: 8

Ref country code: AL

Payment date: 20230718

Year of fee payment: 8